Utilizing cytokines to function-enable human NK cells for the immunotherapy of cancer

Size: px
Start display at page:

Download "Utilizing cytokines to function-enable human NK cells for the immunotherapy of cancer"

Transcription

1 Washington University School of Medicine Digital Open Access Publications 2014 Utilizing cytokines to function-enable human NK cells for the immunotherapy of cancer Rizwan Romee Washington University School of Medicine in St. Louis Jeffrey W. Leong Washington University School of Medicine in St. Louis Todd A. Fehniger Washington University School of Medicine in St. Louis Follow this and additional works at: Recommended Citation Romee, Rizwan; Leong, Jeffrey W.; and Fehniger, Todd A.,,"Utilizing cytokines to function-enable human NK cells for the immunotherapy of cancer." Scientifica.2014, (2014). This Open Access Publication is brought to you for free and open access by Digital It has been accepted for inclusion in Open Access Publications by an authorized administrator of Digital For more information, please contact

2 Scientifica, Article ID , 18 pages Review Article Utilizing Cytokines to Function-Enable Human NK Cells for the Immunotherapy of Cancer Rizwan Romee, Jeffrey W. Leong, and Todd A. Fehniger Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA Correspondence should be addressed to Todd A. Fehniger; Received 20 February 2014; Accepted 2 May 2014; Published 25 June 2014 Academic Editor: Jianhua Yu Copyright 2014 Rizwan Romee et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Natural killer (NK) cells are innate lymphoid cells important for host defense against pathogens and mediate antitumor immunity. Cytokine receptors transduce important signals that regulate proliferation, survival, activation status, and trigger effector functions. Here, we review the roles of major cytokines that regulate human NK cell development, survival, and function, including IL-2, IL-12, IL-15, IL-18, and IL-21, and their translation to the clinic as immunotherapy agents. We highlight a recent development in NK cell biology, the identification of innate NK cell memory, and focus on cytokine-induced memory-like (CIML) NK cells that result from a brief, combined activation with IL-12, IL-15, and IL-18. This activation results in long lived NK cells that exhibit enhanced functionality when they encounter a secondary stimulation and provides a new approach to enable NK cells for enhanced responsiveness to infection and cancer. An improved understanding of the cellular and molecular aspects of cytokine-cytokine receptor signals has led to a resurgence of interest in the clinical use of cytokines that sustain and/or activate NK cell antitumor potential. In the future, such strategies will be combined with negative regulatory signal blockade and enhanced recognition to comprehensively enhance NK cells for immunotherapy. 1. Introduction This review focuses on our current understanding of cytokine-cytokine receptor interactions on human NK cells andhowthesesignalsmightbeusedtopromoteantitumor immunity by NK cells. A brief introduction provides the framework for discussing the impact of cytokines on NK cells and for highlighting the salient features of NK cell biology for effective antitumor responses NK cell development, subsets, education/licensing, target recognition, trafficking, and effector functions. We discuss the cytokine biology of IL- 2, IL-15, IL-12, IL-18, and IL-21 related to NK cells, as well as their translation to the clinic as antitumor immunotherapy. We also highlight a relatively new concept in NK cell biology, innate NK cell memory. As the first form of innate memory directly translated into cancer immunotherapy clinical trials, we focus in depth on cytokine-induced memory-like (CIML) NK cells. Importantly, utilizing cytokines to enhance NK cell functionality is only one part of a comprehensive approach to enhance NK cell antitumor activity, with others including blockade of inhibitory signals/cells, and enhancement of NK cell recognition of tumor target cells (Figure 1). The future of NK cell based therapeutics will involve manipulation of all three intertwined aspects of NK cell biology Human NK Cells. NK cells were originally identified based on their ability to kill tumor target cells in the absence of prior sensitization [1, 2], distinguishing them from adaptive T cells. Over the past 4 decades, it has become clear that NK cells perform more functions than natural killing and participate in multiple ways during host immune defense. Human NK cells are defined phenotypically by the presence of CD56 and lack of T and B cell specific markers (CD3/TCR and CD19) and comprise 5 20% of peripheral blood lymphocytes in normal individuals [3]. Morphologically, resting human NK cells have been identified as large granular lymphocytes, although this description reflects the major CD56 dim NK cell subset in peripheral blood, while CD56 bright NK cells are small lymphocytes. The NK cell activating receptor NKp46 (Ncr1) is highly specific

3 2 Scientifica (3) Block negative regulators/ inhibitors of activation KIR and other inhibitory receptors MHC class I Anti-KIR mabs Anti-NKG2A mabs Anti-PD-1 mabs Treg depletion strategies Block TGF-β Cytokine receptors NK Target cell Activating receptors Activating ligand (2) Utilize cytokines or drugs that augment NK cell functionality Cytokines CIML NK cells Immunomodulatory drugs Exposure to selected viruses (1) Enhance activating receptor signals/triggering KIR haplotype selection KIR: KIR ligand MM Monoclonal antibodies Bi- and trispecific engagers Chimeric antigen receptors Figure 1: General strategy to optimize NK cell immunotherapy. A three-tiered approach to comprehensively modify NK cells for optimal antitumor responses. (1) Enhance NK cell recognition and triggering while providing enhanced specificity, (2) augment functional status using cytokines, immunomodulatory drugs, or prior viral infection, and (3) remove inhibitory signals that include inhibitory KIR/NKG2A/PD-1, block Treg mediated regulation, and block NK cell suppressive cytokines. for NK cells, providing an additional marker that is used in both humans and mice to clearly identify NK cells [3 5]. NK cells are further defined by their functional attributes, including proliferation, production of cytokines/chemokines, natural killing, lymphokine-activated killing, and antibodydependent cellular cytotoxicity (ADCC) via CD16/FcγRIIIa [6].Theyarefoundinmosttissuesinthebodybutare enriched in the spleen, blood, bone marrow, liver, and lymph nodes [7]. This lymphocyte lineage represents one of the first members of a diverse set of recently defined innate lymphoid cells (ILCs) that have distinct transcription factor requirements and differing roles in normal physiology and host defense [8]. The clinical importance of NK cells to normal host defense in humans has been demonstrated in patients that are selectively deficient in NK cells, who develop recurrent, often fatal, viral infections [9]. Further, in rare immunodeficient patients who lack T and B adaptive lymphocytes, NK cells are able to mount an effective antiviral response to cytomegalovirus [10]. In addition, one large epidemiologic study found that low NK cell cytotoxicity predicted an increased risk of developing cancer, suggesting a role in cancer immunosurveillance in humans [11]. Moreover, therapeutic monoclonal antibodies used in cancer patients often mediate their actions via ADCC, with NK cells one effector cell important for such cytotoxicity [12]. More recently, allogeneicnkcellshavebeenutilizedtoinducecancerremissions and participate in the graft-versus-leukemia effect vital to effective allogeneic hematopoietic stem cell transplantation [13, 14]. Thus, the normal properties of NK cells may be harnessed as antitumor immunotherapy strategies [15] NK Cell Development and Education. Human NK cells develop from progenitors in the bone marrow and complete their differentiation and maturation in peripheral organs, especially lymphoid tissues [16, 17]. A number of human NK cell differential intermediates have been identified, most readily isolated from tonsil, lymph nodes, or bone marrow [18 20]. NK cells are thought to become tolerant of normal host tissues through an education process that occurs during development termed licensing [21, 22] but exhibits plasticity depending on the environment the NK cell resides [23, 24], with most of the key aspects of this process defined primarily in model organisms. This results in mature and functional NK cells that are inhibited by germline encoded receptors that recognize self-mhc or related ligands and can effectively sense the loss of such self-ligands or increased expression of activating receptor ligands [25, 26]. A subset of CD56 dim NK cells identified in humans, which fail to express KIR that recognize self-hla ligands, was found to be hypofunctional, suggesting a lack of education and anergy [27]. A number of cytokines have also been implicated in promoting different stages of NK cell progenitor, precursor, and mature NK cell differentiation and survival, especially early acting kit ligand, flt3 ligand, and later acting IL-15 [3, 16, 28, 29]. Cytokines are also responsible for supporting

4 Scientifica 3 NK cell homeostasis, similar to other hematopoietic lineages. As expanded on below, cytokines may also influence the activation state of NK cells, providing a microenvironmentbased cue to augment or diminish the threshold required for triggering NK cells through surface receptors, for example, in the setting of inflammation or upon exogenous provision as immunotherapy [6, 30 34]. Each of these processes is crucial to understand, since the ability to enhance NK cell triggering, augment functionality, and enhance homeostasis may inform translational NK cell based immunotherapy NK Cell Receptor-Based Recognition of Targets. NK cells do not express a dominant, clonally rearranged antigen specific receptor, clearly differentiating this lymphocyte lineage from adaptive T and B cells. Instead, during surveillance NK cells integrate signals from a diverse set of germline DNA encoded activating and inhibitory cell surface receptors [24, 35 37]. In humans, NK cell receptors include killer cell immunoglobulin-like receptors (KIR), C-type lectins (CD94/NKG2A/NKG2C), natural cytotoxicity receptors(ncr;nkp44,nkp30,andnkp46),cd16/fcγriiia, NKG2D, and integrin/adhesion molecules. Best studied, the KIR genes are highly polymorphic and segregate independently from MHC class I, and KIR genotypes impact on NK cell antitumor responses [13, 38]. When inhibitory MHC class I ligands are lost or absent (missing self), inhibitory KIR are not engaged, reducing the signal threshold for triggering. Further, when activating ligands are increased (induced or abnormal self), activation predominates and in some circumstance results in NK cell triggering without loss of MHC class I [39, 40]. When deciding whether to respond to a target cell or not, these signals are combined, including integration of the NK cell activation status influenced by cytokine priming [30, 31, 41, 42] or other events, such as latent viral infection [43]. Recently, prior exposure to CMV and Hantavirus infection has been linked to altered populations of human NK cells, resulting in an expanded NKG2C + NK cells that exhibit enhanced functionality upon restimulation [44 46]. While not a focus of this review, such prior experience of CMV results in a functionally enhanced NKG2C + NK cell population in solid organ or bone marrow transplantation patients [44, 45]. Once appropriately triggered, the NK cell responds by killing the target [47] and producing cytokines including IFN-γ, TNF-α, GM-CSF, MIP-1α, and others[3]. Thus, the activation of NK cells is complex, with influences provided by germline encoded receptors interacting with targets (such as KIR) and also NK cell s microenvironment and prior exposure to viruses and cytokine receptor signals, all of which provide translational opportunities Human NK Cell Subsets. Two phenotypically and functionally distinct subsets of NK cells have been well defined in human peripheral blood [48 53]. CD56 bright NK cells are numerically minor subset, comprising 1 15% of the total NK cell population in blood. These NK cells have no or low CD16 expression, generally lack KIR receptors, express inhibitory CD94/NKG2A, are poorly cytotoxic at rest, express distinct chemokine and cytokine receptors, and prefer secondary lymphoid tissue, compared to CD56 dim NK cells. This subset of NK cells has been implicated in a wide variety of physiologic roles in health and disease, including production of effector cytokines in response to accessory cell cytokines [51], control of EBV [54], immunoregulation [55], and networking between adaptive and innate immunity [50, 56]. CD56 dim NK cells express high levels of CD16 (CD16 bright ) and mediate ADCC, contain abundant cytotoxic granules loaded with perforin/granzyme, express KIR [51], and respond more robustly to surface-receptor mediated activation when interacting with potential target cells [57, 58]. CD56 dim NK cells can be further subdivided into less mature (CD94+NKG2A+CD57 ) and more mature (CD94 NKG2A CD57+NKG2C+) subsets [59 61]. Thus, due to CD56 dim NK cell abundance and their functional properties, this subset has been the focus of most studies evaluating anticancer properties of NK cells. More recent evidence suggests that cytokine priming allows for robust antitumor response by CD56 bright NK cells, opening up the possibility that this NK cell subset is also significant for immunotherapy [62]. It has been reported that CD56 bright NK cells may differentiate into CD56 dim NK cells [63 65]. However, in vitro differentiation systems from CD34 + hematopoietic progenitors typically yield CD56 bright type NK cells with a lower percentage of NK cells with CD56 dim attributes (e.g., KIR and CD16), and patients with mutant GATA2 have a selective loss of CD56 bright NK cells [66]; thus it remains plausible that these two NK cell subsets have distinct ontogeny. Regardless of their direct development relationship, each subset also clearly retains its own unique biology in the form of receptor repertoires, preferred modes of stimulation, tissue localization, and primary effector functions and should be evaluated in the context of NK cell responses to malignancy. 2. How NK Cells Functionally Contribute to Antitumor Immunity 2.1. NK Cell Cytotoxicity. Triggering resting NK cells to kill occurs through integration of activating and inhibitory receptor signals, which is referred to as natural killing [35]. Stimulation with IL-2 or IL-15 for several days results in lymphokine-activated killer (LAK) cells, which have the capacity to kill additional targets that are resistant to resting blood NK cells [67]. For human NK cells, CD16 (the FcγRIIIa receptor) is a major activating receptor that recognizes antibody-coated target cells, and killing through this activation mode is referred to as antibody-dependent cellular cytotoxicity (ADCC) [3]. An NK cell utilizes two main mechanisms to kill tumor cells, following these triggering recognition events: granule exocytosis and death receptors. For granule exocytosis, cytotoxic granules that contain perforin, granzymes, and other effector proteins of cytotoxicity arereleasedintoatightcytotoxicsynapse[47, 68]. Perforin facilitates granzyme entry into the target cells, where these serine proteases cleave targets to induce an apoptotic-like cell death. A second pathway for inducing cell death is using cell

5 4 Scientifica surface receptors, most commonly Fas ligand and TRAIL. Finally, secreted TNF-α and IFN-γ may induce a senescent tumor cell death, especially when coordinately secreted [69]. Importantly, activation through cytokine receptors may augment all of these mechanisms of NK cell killing NK Cell Cytokine Production and Immune Networking. One major function of NK cells is production of cytokines and chemokines following either cytokine- or activating receptor stimulation on the NK cell surface. The prototype effector cytokine produced by NK cells is IFN-γ, which has pleotropic effector actions on other immune cells, antigen presenting cells, and virally infected or malignant target cells. Additional cytokines (GM-CSF and TNF-α) and chemokines (MIP-1α, MIP-1β, and RANTES) are also produced, which depends on the NK cell stimulation type and the time course after activation [6]. Through cytokines NK cell may impact other immune responders, including T cells, and influence adaptive immunity by activating antigen presenting cells [34]. Indeed, it is thought that NK cells participate in a complex interaction network with other lymphocytes, dendritic cells, and macrophages to effectively control infection. For antitumor immunity, NK cells may promote Th1 type T cells responses, activate antigen presenting cells, and induce tumor cell death to facilitate antigen presentation. Thus, immunotherapy approaches should not only focus on the ability of NK cells to degranulate and kill tumor targetsinvitrobutalsoenhanceantitumorimmunitythrough such indirect mechanisms. This remains a challenge in the evaluation of human NK cell antitumor responses in patients, sinceitisnotclearthatsamplingtheperipheralbloodyields the most relevant information to infer activities at the site of the tumor or elsewhere such as secondary lymphoid organs. While such studies are technically (and financially) challenging and require careful consideration of potential risks by physicians and patients, expanded sampling including the site of the tumor/metastasis and lymphoid organs would be highly informative for a more complete picture of NK cell responses in vivo. 3. General Concepts: Cytokine Receptors on Human NK Cells Cytokine receptors are important for a wide variety of NK cell events, including development, proliferation, homeostasis, and activation status key aspects of biology for immunotherapy. Human NK cells have been shown to constitutively express a number of cytokine receptors, which transduce signals when ligated through a number of intracellular signaling pathways (Table 1). In addition, selected cytokine receptors or subunits may be induced or their expression may be enhanced upon activation, providing one mechanism of synergy between different cytokines. Further, it is known that combined signals through multiple types of cytokine receptors, or cytokine plus activating NK cell receptors, result in the most robust NK cell effector responses. In this fashion, NK cells may sense differing extents of inflammation and respond with a continuum of intensity, providing a mechanism for tuning the extent of a response to the pathogenic situation. For example, IFN-γ is produced at very low amounts when IL-2/IL-15, IL-12, or IL-18 receptors are individually activated; however, with combinatorial stimulation there is a dramatic, cytokine dose-dependent, and synergistic effect on NK cell IFN-γ secretion [70]. While challenging to definitively address via experimentation, this maybemostrelevantinvivowhencytokineconcentrations are limiting, and therefore NK cells are exposed to suboptimal cytokine receptor stimulation. Further, cytokinebasedsignalsmayalsoaltertherulesforreceptor-based licensing, for example, in the setting of ongoing infection or inflammation [71], an area that is relatively unexplored in NK cell responses to tumors. While negative cytokine regulation of NK cell activation is not a focus of this review, there are clear examples where anti-inflammatory cytokines turn off NK cells, such as TGF-β that rapidly inhibits multiple aspects of NK cell functionality [72]. In some situations including the tumor microenvironment, TGF-β effects may be reversed, suggesting that inhibitory cytokine blockade may be feasible as an approach to enhanced NK cell responses [73]. NK cell cytokine receptors activate a wide variety of intracellular signaling pathways, providing one mode of cooperation and a method to separate induction of different NK cell functional programs. The role of IL-2, IL-15, IL-12, IL-18, and IL-21 in human NK cell biology is reviewed in the following sections, with emphasis on newer findings, followed by translational studies in cancer patients (Table 2). It is important to note that investigation of human NK cells in vivo is difficult and that early phase clinical trials provide a useful platform to advance our knowledge of how cytokines impact human NK biology in health and disease. 4. IL-2 and IL-15: Basic Biology IL-2 and IL-15 represent the best studied cytokine activators of NK cells and have a number of positive functional effects on NK cells to enhance antitumor responses [28, 29, 74, 75]. Signals downstream of the IL-2/15R have been extensively characterized and include activation of the Jak1/3 and STAT3/5, the PI3K pathway, the MAPK pathway, and ultimately NF-κB. These signals through the IL-2/15R are central for NK cell development and homeostasis, induce proliferation, costimulate cytokine production, and enhance cytotoxic effector mechanisms [28, 29, 74, 75]. IL-2 and IL-15 share the IL-2/15Rβ and γ c as the primary signaling subunits and interact with this heterodimer with intermediate affinity (IA), requiring nanomolar concentrations. CD56 bright NK cells constitutively express CD25/IL-2Rα, which forms a high affinity heterotrimeric IL-2Rαβγ that responds to picomolar concentration of IL-2 [76, 77]. CD25 and the high affinity (HA) receptor can be induced on both CD56 bright and CD56 dim NK cells following combined cytokine activation with IL-12+IL-15+IL-18 and to a much lesser extent following IL-2 or IL-15 activation [78 80]. The IL-15Rα is primarily expressed on dendritic cells and macrophages, has high affinity for IL-15 as a single subunit, and transpresents bound

6 Scientifica 5 Table 1: Cytokine receptors on human NK cells. Receptor Components NK subset Signaling Functions Source IL-2R IL-12R IL-15R IL-18R IL-21R IL-2Rα IL-2/15Rβ γ c IL-12Rb1 IL-12Rb2 IL-15Rα IL-2/15Rβ γ c IL-18Rα/R1 IL-18Rβ/RAP IL-21R γ c IL-2Rαβγ: CD56 bright IL-2Rβγ: CD56 bright and CD56 dim CD56 bright and CD56 dim CD56 bright and CD56 dim CD56 bright and CD56 dim CD56 bright and CD56 dim Jak1/3, STAT3/5 PI3K Ras/Raf/MAPK Jak2/Tyk2 and STAT4 Jak1/3 and STAT3/5 PI3K Ras/Raf/MAPK MyD88, IRAK4/TRAF6, MAPK, and NFkB Jak1/3 and STAT1/3/5 Cytokine production Proliferation Survival Enhanced cytotoxicity Cytokine production Enhanced cytotoxicity Cytokine production Proliferation Survival Enhanced cytotoxicity Cytokine production Proliferation Enhanced cytotoxicity Enhanced ADCC Cytokine production Limits proliferation Tcell DC Mφ DC Mφ BM stroma DC Mφ Tcell IL-15 to the IL-15Rβγ c complexonnkcells[29, 75]. IL- 2isproducedbyactivatedTcells,andIL-2Rαβγ has been showntofacilitatecytokine-basedcrosstalkwithtcells[50], especially during an active immune response. IL-15Rα/IL- 15 is produced by a number of APCs and likely represents the central pathway whereby IL-2/15R signals are triggered during normal physiology [34]. This is supported by the NK cell phenotypes of the relevant knockout mice: IL-2 / and IL-2Rα / have an intact NK cell compartment, whereas IL- 15Rα / and IL-15 / mice have a marked (>20-fold) reduction in NK cells [28, 75]. In vitro, nanomolar concentrations of IL-2 or IL-15 both activate the IL-2/15Rβγ c and have similar functional effects. In vivo, it is critical to parse whether IL-2 doses are low (picomolar) and only sufficient to ligate IL-2Rαβγ or intermediate/high (nanomolar) which can ligate the IA IL-2/15Rβγ c. IL-2 has been extensively studied in cancer patients [29, 74] and overall yielded unexpectedly few clinical responses as a single agent, likely due to simultaneous induction of regulatory T cells, which can limit NK cell responses. Recently, IL-15 has entered clinical trials and has promised to modulate NK cells (and effector T cells) in the absence of Treg induction. These cytokines are also used in ex vivo activation and/or expansion of NK cells for adoptive immunotherapy and to support the expansion and function of NK cells after infusion. 5. IL-2 and IL-15: Translation IL-2 was one of the first cytokines used clinically, with hopes of inducing antitumor immunity. The clinical use of recombinant human (rh)il-2 has been reviewed extensively [29, 81] (Table 2). As a single agent at high dose IL-2 induces remissions in a minority of patients with renal cell carcinoma (RCC) and metastatic melanoma, with an unclear mechanism of action [81]. Presumably, ligation of the IL-2/15Rγβ c on immune cells is contributing to the clinical activity and substantial toxicity associated with this rhil-2 dose. A detailed characterization of the effects of high dose IL-2 on the NK cell compartment in vivo has not been reported. Following the experience with high dose IL-2, low dose IL-2 was investigated and aimed at selectively ligating the HA IL-2Rαβγ in an effort to reduce toxicity while maintaining biological activity [82]. At the time, these studies resulted in the expansion of CD56 + CD3 NK cells ( % increase) predominantly from the CD56 bright CD16 + NK cell subset [83 87]. While these NK cells were active against NK cell sensitive targets (K562) and could mediate ADCC, they required additional stimulation with high dose IL-2 to mediate LAK activity against NK cell resistant tumors. This was consistent with the serum levels of IL-2 achieved in vivo of picomolar, indicating selective stimulation of IL-2Rαβγ. Later studies suggested that low dose IL-2 facilitated CD56 bright NK cell differentiation from progenitors and survival in vivo, with minimal impact on peripheral proliferation [88]. Additional studies explored low dose IL-2 therapy in combination with intermediate dose pulses to first expand and then activate NK cells in vivo, which effectively enhanced NK cell function in vivo [86]. This approach was also combined with antitumor monoclonal antibodies, which appeared safe and resulted in some clinical responses in early phase studies [89]. Subsequent to these clinical studies, the biology and the central role of IL-2 for regulatory T cell homeostasis and function were defined [90]. Follow-up studies demonstrated that low dose IL-2 therapy also expanded regulatory T cells, which are known to limit NK cell responses, in addition to effector T cells [91 93].

7 6 Scientifica Cytokine IL-2 IL-2 IL-2 Additional agent Post NK cell infusion Post NK cell infusion Rituximab and NK cell infusion Table 2: Summary of selected cytokine clinical trials and major findings. Disease Major biologic effects Outcomes Reference AML, HL, RCC, and melanoma AML CD20 + NHL In vivo activation and expansion of the NKcellsinsomeofthepatients Minimal expansion of the adoptively transferred NK cells Preferential expansion of recipient regulatory T cells IL-12 None RCC Profound increases in serum IFN-γ IL-12 None Melanoma IL-12 IL-18 IL-18 IL-18 IL-21 None Rituximab None Rituximab None RCC, melanoma, CC, and others RCC, melanoma, and HL Advanced melanoma and RCC Advanced CD20 + NHL Metastatic melanoma Transient decrease in CD8 + and CD16 + lymphocytes in peripheral blood and neutrophils along with high serum levels of IFN-γ and IL-10 Transient decrease in T cells, B cells, and NK cells. Transient increase in the expression of CD2, CD11a, and CD56 on NK cells. Increase in the cytotoxic activity of the NK cells Transient decrease in lymphocytes (CD4 +,CD8 +, NK, and NKT cells, but most profoundly in the NK cells). Increased Fas ligand on NK cells, CD8 + T cells, and NKT cells. Increased serum levels of IFN-γ, GM-CSF, IL-18 binding protein, and soluble Fas ligand. Some patients (38%) developed antibodies to rhil-18 Transient lymphopenia (CD4 + Tcells, CD8 + T cells, and NK cells) which correlated with the expression of CD69. Increased plasma concentrations of INF-γ, GM-CSF, TNF-α,CXC chemokine IP-10, and CC chemokine MCP-1. Antibodies against hril-18 developed in 32% of the patients Transient lymphopenia with undetectable circulating B cells. Increase in plasma concentrations of IFN-γ, GM-CSF, TNF-α,MIG,IP-10,andMCP-1 Transient increase in the serum levels of scd25 (an immune activation marker). Perforin-1 and granzyme B expression in CD8 + T cells and NK cells. Increase in cytotoxic activity of the NK cells CR in some of the AML patients Prolonged persistence of CR in some patients Induction of CR in some patients No major responses noted; phase II part terminated due to major toxicities including 2 deaths Decrease in the size of tumors in some patients No major responses with IL-12 therapy reported in this trial Partial response in few patients No major responses reported though the drug was well tolerated without any major side effects Overall response rate of around 26%, complete response rate of 11%, and partial response rate of 16% Response seen only in one patient [153] [154] [156] [109] [110] [111] [166] [123] [125] [124] [135]

8 Scientifica 7 Cytokine IL-21 IL-21 IL-21 Additional agent None Cetuximab None Table 2: Continued. Disease Major biologic effects Outcomes Reference Metastatic melanoma Metastatic colorectal carcinoma Metastatic melanoma Increased serum levels of scd25 and increased CD25 expression on NK cells and CD8 + T cells. Increased expression of IFN-γ, perforin, and granzyme B in NK cells and CD8 + Tcells Decreased number of NK cells, CD8 + T cells, and B cells. Increase in the cytotoxic activity of NK cells. Increase in the absolute number and the expression of CD64 (FcγRI). Increased serum levels of scd25 Increase in the serum scd25 levels Clinical response seen only in two patients Stable disease was reported in 60% of the patients Overall response rate was 23% and median overall survival of 12.4 months in these patients compared favorably with a median survival of 8.4 months predicted from historic controls This table included IL-2 provided in the context of allogeneic NK cell infusions, while early studies of IL-2 have not been included in this table and have previously been reviewed [29], [74], [81]. [136] [138] [137] Indeed, low dose IL-2 has recently been more studied to control immune-based diseases through Treg augmentation, such as graft-versus-host disease. Thus, while initial studies suggested CD56 + CD3 NK cell functional modulation, the potential for antitumor immunity was likely limited by Treg expansion, which remains a concern for NK cell adoptive transfer approaches that use rhil-2 postinfusion to support NK cells. While rhil-2 is routinely administered to patients following NK cell adoptive immunotherapy, modulation of regulatory T cells is a substantial concern for this practice, andifusedcombinationswithanti-tregtherapyarelikely warranted. Further, alternative cytokines (such as IL-15) that do not augment Treg number and function should be explored. IL-15 was initially viewed as very similar to IL-2 in its cytokine biology, and while its ability to stimulate NK cell development, homeostasis, and functionality was remarkable, it was not initially pursued with rhil-2 already clinically available [28, 29, 74]. Clinical interest in IL-15 was rekindled when the Treg effects of rhil-2 were unraveled, and the distinct IL-15 receptor biology was reported [94]. Based on effects of both T and NK cells, rhil-15 (in the absence of the IL-15Rα) is under clinical investigation in solid tumors (melanoma, renal cell carcinoma: NCT , NCT ; advanced cancers NCT , NCT ) andtosupportnkcellsafter adoptive transfer in leukemia patients (NCT ). Studies performed in nonhuman primates at the NIH administering subcutaneous rhil-15 intermittently every 3 days demonstrated low toxicity with expansion of NK cells (in addition to CD8 memory and CD4+ T cells) in the absence of Treg expansion in vivo [95]. Interestingly, daily administration for 14 days resulted in reversible toxicities in two macaques consisting of neutropenia with a hypocellular bone marrow and anemia with a lymphoid infiltrate in the bone marrow, coinciding with a marked peripheral lymphocytosis. Of note, plasma concentrations were sustained with 15 mcg/kg rhil-15 predose levels of pg/ml and peak levels of pg/ml. Pharmacokinetic analysis of interrupted doses indicated clearance of rhil-15 prior to each dose (<10 pg/ml), with peak levels variable ( pg/ml) depending on the IL-15 dose (2.5, 5, or 10 mcg/kg) administered. Preliminary reports from a phase 1 study of rhil-15 in AML patients following an allogeneic NK cell infusion suggest that rhil-15 given as an IV bolus 3 times weekly results in allogeneic NK cell expansion, and the expanded NK cells are functional [96]. The results of these initial clinical trials will inform future approaches as an NK cell and antitumor immunity modulator. Thus, IL-15 remains highly interesting as an NK cell modulator for immunotherapy but will likely have non-nk cell immune effects that will require close monitoring, with the potential for toxicity distinct from rhil-2. Further, there is the potential to simultaneously augment both NK cell and T cell functions, which may result in crosstalk that further enhances antitumor immunity, compared to modulation of NK cells or T cells in isolation. This cytokine was identified as a top priority

9 8 Scientifica bythencisponsoredimmunotherapyagentworkshop ( NCI Workshop Proceedings 23Oct07.pdf), but currently access remains limited for clinical trial investigation. Since IL-15 requires IL-15Rα for efficient ligation of the IL-2/15Rβγ c in vivo, several studies evaluated coadministration of IL-15/IL-15Ra complexes on NK cells [97, 98]. These resulted in enhanced in vivo activity, and the use of IL-15/IL-15Rα complexes or fusion proteins remains highly promising as an IL-15 immunotherapy. One approach uses fusion of an IL-15 mutein with higher affinity, coupled to the IL-15Rα sushidomain,fusedtoanfcdomainto stabilizethecomplex(alt-803)[99, 100]. This results in an IL-15 mimic with prolonged in vivo half-life that selftranspresents to IL-2/15Rα [100]. Preclinical studies of ALT- 803 are promising with prolonged in vivo persistence after a single injection, and early stage clinical trials are in progress to evaluate safety and immunomodulation in advanced melanoma (NCT ) and relapsed malignancies after allogeneic SCT (NCT ) using a weekly schedule. A detailed comparison of ALT-803 to rhil-15 for human NK cell stimulation and function has not been reported. It is likely that the dose of ALT-803 will be critical, considering its pharmacokinetics, to avoid the potential toxicity observed with daily rhil-15 in macaques. If effective and safe, this approach would provide a more practical administration approach for IL-15 and the potential for expanded clinical trials to combine with monoclonal antibodies or NK cell infusions for a variety of malignancies. 6. IL-12 Basic Biology IL-12 is a heterodimeric cytokine composed of p35 and p40 subunits (IL-12α and β chains), originally identified as NK cell stimulatory factor (NKSF) based on its ability to enhance NK cell cytotoxicity [101]. Upon encounter with pathogens, IL-12 is released by activated dendritic cells and macrophages and binds to its cognate receptor, which is primarily expressed onactivatedtandnkcells[102]. Subsequent dimerization of the IL-12Rβ1 and IL-12Rβ2 subunits of the IL-12 receptor transducessignalsthroughjanusfamilykinases(jak2and TYK2) and STAT family members, including STAT3, 4, and 5[101]. The primary effects of IL-12 on NK cells, including IFN-γ and TNF-α production, have primarily been attributed to STAT4-mediated signaling. Initial studies provided evidence that rhil-12 augmented human NK cells cytotoxicity and proliferation [103]. Notably, the IL-12R is expressed on resting NK cells, thereby facilitating rapid immune responses without prior activation [104]. Evidence provided by in vitro and in vivo studies of synergism between IL-12 and other activating stimuli suggests that IL-12 likely acts on NK cells in concert with other cytokines, such as IL-2 and IL-18, or with receptor-based interactions from pathogenic cells [104, 105]. 7. IL-12 Translation Numerous preclinical studies suggested that IL-12 had antitumor potential, including IFN-γ dependent antitumor responses against melanoma and renal cell carcinoma (RCC) cell lines in mice [106, 107]. In the first phase I study Atkins et al. used a regimen with a 2-week rest period between first and subsequent intravenous bolus doses of rhil-12 and determined 500 ng/kg as the maximal tolerated dose (MTD) [108]. Biologic effects in these patients with advanced malignancies included dose-dependent increases in IFN-γ and IL-12-induced lymphopenia in the peripheral blood (T and NK cells). In this study few patients had tumor response and some patients had stable disease and therefore demonstrate safety and potential clinical utility of this cytokine. However, a subsequent phase II study by the samegroupinpatientswithadvancedrenalcellcarcinoma IV bolus dose of 500 ng/kg of rhil-12 was associated with severe toxicities and two patients died [109]. This phase II study had not used a rest period between first and subsequent consecutive daily dosing. This alteration in the rhil-12 regimen was thought to be responsible for causing these severe toxicities as the use of single IL-12 injection prior to subsequent consecutive daily dosing protected mice and cynomolgus monkeys from acute toxicity [109]. In a pilot study Bajetta et al. treated 10 metastatic melanoma patients with fixed dose of rhil-12 (0.5 μg/kg) on days 1, 8, and 15 [110]. This regimen was overall well tolerated with few patients demonstrating reduced tumor size. In another study, Robertson et al. used an IV bolus regimen of rhil-12 in patients with advanced malignancies and found transient increase in the cytotoxic activity and expression of CD2, CD11a, and CD56 on NK cells, in addition to causing transient lymphopenia which was particularly marked for NK cells [111]. No major responses were seen in this study of refractory solid tumor patients. In a phase II randomized control trial, Motzer et al. compared rhil-12 with interferon-α (IFN-α) in patients with previously untreated but advanced renal cell carcinoma [112]. In this study rhil-12 was given subcutaneously and was well tolerated but trial was terminated before completion duetolowresponseratesintherhil-12arm.morerecent preclinical studies have highlighted the potential for IL-12 to costimulate NK cell IFN-γ production in combination with antitumor monoclonal antibodies [105, 113, 114]. These findingshavebeentranslatedintoseveralphase1and2 clinical trials of IL-12 administered in concert with antitumor monoclonal antibodies in head and neck carcinoma [115, 116] and lymphoma[117]. Importantly, correlative studies in one trial associated NK cell IFN-γ production ex vivo with clinical responses. Addition of IL-12 to rituximab resulted in a modest response rate (37%), while provision of IL-12 after progressiononsingleagentrituximabfailedtoinduceany responses [117]. Anongoingmulticentertrialisinvestigating subcutaneous rhil-12 administered in combination with cetuximab for patients with relapsed/refractory head and neck cancer (NCT ). Basedupontheearlyinflammatorytoxicityandminimal response rates observed in several different early phase studies, commercial clinical development of rhil-12 as a single agent is unlikely. However, several more recent early phase studieshaveidentifiedadoseandascheduletocombine IL-12 with antitumor monoclonal antibodies, suggesting that safe administration is possible and modulation of NK cell in

10 Scientifica 9 vivo is achieved. Future strategies could alternatively include fusing IL-12 to an antitumor monoclonal antibody, potentially in the context of trispecific reagent that also engaged CD16, which would allow combined IL-12/CD16-mediated activation of NK cells at a tumor site. Based primarily on its potential as a vaccine adjuvant, IL-12 was also identified as a priority cytokine for clinical development by the NCI Immunotherapy workshop; however, access for new clinical trials remains limited. 8. IL-18 Basic Biology IL-18 is a member of the proinflammatory IL-1 family and, like IL-12, is secreted by activated phagocytes [118]. In contrast to other cytokines that transduce signals through the JAK-STAT pathway, the IL-18 receptor (IL-18R) primarily transduces signals through the adapters MyD88 and TRAF6 leading to MAP kinase and NF-kB activation, although minor activation of STAT3 has been reported [119]. In NK cells, IL-18 has traditionally been described as a costimulatory cytokine that functions synergistically with IL-12 and IL- 15 [70, 120], particularly because IL-12 signaling upregulates IL-18R expression in T cells [121]. However, the IL-18Rα is constitutively expressed on unstimulated NK cells and can induce NK cell proliferation alone, although the addition of IL-15 greatly enhances proliferation [120]. Other distinct roles for IL-18 include reports that dendritic cell-derived IL-18 primes NK cells to produce more IFN-γ during later stimulation [42]. Additionally, NK cells from IL-18 deficient mice have impaired cytotoxicity and IFN-γ production, indicating the importance of this cytokine to NK mediated host defense [122]. 9. IL-18 Translation Despite having demonstrated significant antitumor activity in preclinical animal models, rhil-18 has been studied in only few clinical trials to date [ ]. Robertson et al. in a phase I study demonstrated the relative safety of using rhil-18 in patients with advanced malignancies but no major clinical responses were seen in these patients [123]. In another study by the same group, patients with advanced metastatic melanoma and renal cell carcinoma were treated with escalating doses of rhil-18 [125]. In this study doses as high as 2,000 μg/kg bw were relatively well tolerated but again no major responses were seen. In a recent study, rhil-18 was used in combination with rituximab for patients with CD20 + B cell non-hodgkin s lymphomas to potentially augment the ADCC function induced by rituximab [124].Again no dose limiting toxicity was observed when used in combination with rituximab and some responses were seen in 5 patients (2 complete and 3 partial responses). In all of these studies, rhil- 18 administration led to transient lymphopenia along with markers of NK cell activation like increased plasma levels of IFN-γ, TNF-α, and GM-CSF which were observed. rhil- 18 treatment led to the development of antibodies against IL-18 in some of these patients. Currently rhil-18 is being studied in combination with ofatumumab which is a fully human monoclonal antibody against CD20 and known to mediate more potent ADCC against CD20 + lymphoma cells (NCT ). In the absence of tumor responses in early stage clinical studies, continued commercial development of rhil-18 as a single agent drug appears unlikely. 10. IL-21 Basic Biology The IL-21 receptor is predominantly expressed on T, B, and NK cells and binds to IL-21 produced by activated T cells [126]. The IL-21 receptor forms a heterodimer with the common cytokine-receptor γ-chain. Binding of IL-21 to NK cells induces the phosphorylation of STAT1, 3, and 5, although STAT3 has been shown to be the dominant transducer following receptor engagement [127]. Minor engagement of the PI3-kinase and MAP kinase pathways by STAT-independent activation has also been reported [127]. Downstream effects in lymphocytes, including T and B cells, have implicated IL-21 in the negative regulation of cell survival [128, 129], alterations in immunoglobulin isotype switching [130], and promotion of Th17 development [131]. Although IL-21R / mice have normal NK cell numbers, treatment of murine NK cells in vitro with IL-21 reduces NK cell proliferation and survival in the presence of IL-2 or IL-15 but induces terminal differentiation and enhances NK cell cytotoxicity against tumor lines [132]. Further studies have shown that IL- 21 s effects on tumor protection were dependent on NKG2Dmediated recognition of tumor cells by NK cells [133]. 11. IL-21 Translation Due to its ability to stimulate NK cells and CD8 + Tcells, IL-21 is an attractive cytokine for antitumor immunotherapy. For ex vivo NK cell expansion, membrane bound IL-21 has been expressed in K562 stimulator cells, with effective results [134]. Davis et al. demonstrated that rhil-21 was safeandwelltoleratedinaphaseistudyinpatientwith metastatic melanoma [135], although clinical efficacy was limited to one partial response. rhil-21 was also shown to increase soluble CD25 and induce expression of perforin and granzyme B on CD8 + cells. A phase IIa trial in patients with untreated metastatic melanoma also showed safety and minimal adverse events, and several clinical responses were reported [136]. Immune correlative studies showed that rhil- 21 increased soluble CD25 and increased expression of CD25, IFN-γ, perforin, and granzyme B in both CD8 + Tcellsand NK cells. In a recent phase II study by Petrella et al., rhil- 21 treatment of patients with metastatic melanoma induced anoverallresponserateof23%andtheirmedianprogression free and overall survival also favored the historic controls [137].BasedupontherationalethatrhIL-21augmentsADCC function, Steele et al. recently reported the use of rhil-21 in combination with cetuximab in a phase I trial in patients with stage IV colorectal cancer [138]. The combination was well tolerated and 9/15 patients exhibited stable disease with therapy, but the study was prematurely terminated due to a sponsor decision. Analysis of correlative studies in these patients showed not only a drop in blood NK cells but also an increased cytotoxic functionality of NK cells against K562 targets. In vitro studies indicate that IL-21 augments NK cell

11 10 Scientifica (1) Preactivation NK IL-15 Control Cytokines 16 hours wash Rest 1 6 weeks 6 hours in vitro w/il-15 or tumor targets IL-12+ IL-18+ IL-15 Preactivated (2) Rest and return to basal activation state (3) Restimulate Proliferation Survival/persistence CD69 and NKG2D GzmB and CD25/IL-2Rαβγ CD56 CD56 (4) NK cell functions % Control % CIML IFN-γ Cytokine production Cytotoxicity Figure 2: Overview of human cytokine-induced memory-like NK cells. Human NK cells preactivated with IL-12+IL-15+IL-18 for 16 hours return to a basal activation status. Weeks later CIML NK cells have evidence of proliferation and increased expression of CD69, NKG2D, granzyme B, and CD25, compared to control IL-15 preactivated NK cells. When CIML NK cells are restimulated, they exhibit enhanced functional responses, including cytokine production and cytotoxicity against leukemia targets. Similar results are observed when preactivated or control human NK cells are adoptively transferred into immunodeficient NOD-SCID-γ / c mice and evaluated 7 days later for in vivo persistence and enhanced functionality. ADCC against CLL cells in vitro, in addition to direct effects on CLL cells [139]. Clinical studies have explored rhil-21 in combination with rituximab for NHL with a 42% overall response rate, and in vivo activation of NK cells was observed based on the surrogate marker CD69 (NCT ). Thus, rhil-21 appears to have modest clinical effects on solid tumors, favorable toxicity profile, and evidence of biological modulation in vivo in patients. Based on this potential for augmented ADCC, clinical development will likely continue. 12. Cytokine-Induced Memory-Like NK Cells: Basic Biology NK cells are traditionally considered members or the innate branch of the immune system that respond rapidly but lack immunologic specificity in the form of a clonal antigen receptor and memory of prior activation. Recently several groups have challenged this paradigm of NK cells as pure innate lymphocytes and demonstrated memory-like functions in NK cells [ ]. The von Adrian group first reported recall responses in cells exhibiting an NK cell phenotype to haptens during delayed hypersensitivity reactions in Rag1 / mice [141]. The Lanier group demonstrated enhanced function by Ly49H + NK cells upon Ly49H-based restimulation in mice after resolution of an acute MCMV infection [140]. Cooper et al., in the Yokoyama lab, described cytokineinduced memory-like (CIML) NK cell functions in mice defined by initial combined cytokine activation, a subsequent return to the resting state after adoptive transfer in vivo, and an enhanced functionality with later restimulation [142, 144]. Murine NK cells which had previously been activated with IL-12, IL-15, and IL-18 exhibited increased IFN-γ response upon their cytokine restimulation, compared to IL-15 alone, for 1 4 months after adoptive transfer [142]. This enhanced functionality was accompanied by extensive proliferation and passed on following cell division, suggesting a durable change in the NK cell program. This was not simply an alteration in the transcription of the IFN-γ locus or IFN-γ mrna stability, since IFN-γ mrna levels were not different in mouse CIML and control NK cells. Further, CIML NK cell activity persisted after extensive homeostatic proliferation in immunodeficient Rag / γ / c recipients for at least 1 month [145]. Studies from the Cerwenka lab demonstrated that a single infusion of IL-12, IL-15, and IL-18 preactivated NK cells protected against established tumor cell line implant (B16 melanoma and RMA-S lymphoma). In this system, CIML NK cell responses required CD4+ T cell-derived IL-2. More recently, mouse memory NK cells that arise after an acute MCMV infection require proinflammatory cytokines, including IL- 12, that suggests a similar mechanism of differentiation in these two types of innate NK cell memory [146]. Thus, CIML NK cells are able to remember prior activation (at least for months) and persist in the host with an enhanced functional capacity. Ourgroupfirstdemonstratedthatabriefcytokinepreactivation of human NK cells with IL-12, IL-15, and IL-18 or other combinations (e.g., IL-15+IL-18) induces human CIML NK cells (Figure 2)[147]. Following preactivation with

12 Scientifica 11 control (IL-15 only) or CIML-inducing (IL-12+IL-15+IL-18) cytokines for 16 hours, purified NK cells (>95% CD56 + CD3 ) were washed and rested in low dose IL-15 to maintain survival. Human CIML NK cell that rested for 1-2 weeks exhibited increased IFN-γ production, compared to control NK cells, after restimulation with cytokines (IL-12+IL-15) or triggering by leukemia cell targets. In addition, coculture of IL-12, IL-15, and IL-18 preactivated NK cells with other PBMC resulted in long lived (6 weeks) functionally enhanced NK cells. Similar to the murine studies, human CIML NK cells maintained their enhanced functionality following extensive cell division, suggesting a sustained change to key aspects of the NK cell molecular program [147]. Both flow sorted (>98% pure) CD56 bright and CD56 dim human NK cell subsets exhibited a memory-like functionality, although the magnitude of CIML responses appeared modestly higher in CD56 bright NK cells. Several surface markers were increased on CIML NK cells, CD94/NKG2A, CD69, and NKp46, compared to controls. For CD56 dim NK cells, there appeared to be enrichment of memory-like function in NK cells with expression of CD94, NKG2A, NKG2C, and CD69 and those that lacked KIR and CD57. More recent studies have demonstrated that human CIML NK cells express increased granzyme B protein and increased cytotoxic function against leukemia target cells, compared to control NK cells from the same donor. Further, these CIML NK cells responded more robustly to allogeneic AML blasts, suggesting the potential for immunotherapy of AML and potentially other malignancies [148]. From a physiologic viewpoint, human CIML NK cells may arise in the setting of infection or inflammation, and in this situation it is likely that dendritic cells or macrophages are the source of combined cytokine activation. Both human and mouse CIML NK cells have an increased expression of CD25 (IL-2Rα), a key component of the high affinity heterotrimeric IL-2 receptor (IL-2Rαβγ receptor) [149, 150]. For human NK cells [150], this induction is highly robust on both CD56 bright and CD56 dim NK cells after hours and is maintained for at least 1 week. Induction of CD25 results in a signal-competent IL-2Rαβγ, since CIML NK cells exhibit enhanced STAT5 phosphorylation in response to picomolar concentrations of IL-2 compared to controls. Further, picomolar concentrations of IL-2 are able to selectively impact CIML NK cells in vitro via enhanced costimulation of IFN-γ, cytotoxicity against leukemia targets, and proliferation. CIML NK cells also exhibit preferential expansion and maintenance of their enhanced functionality following adoptive transfer into immunodeficient NSG mice with rhil-2 administration. Thus, enhanced responsiveness to IL-2 is another attribute of CIML NK cells and may provide a clue to their interactions with T cells and the ability of exogenous low dose IL-2 (in addition to IL-15) to support their expansion and function in vivo. Recently, mouse NK cells were shown to be modulated by Treg sequestration of IL-2 in vivo [151], and we hypothesize that CIML NK cells, through expression of IL-2Rαβγ at a high density, may more effectively compete for IL-2 than naïve NK cells and may be resistanttothismodeoftregsuppression. Innate NK cell memory is a relatively new field with a lack of studies on the mechanisms that underlie their differentiation. For CIML NK cells, while the cell biology has been functionally characterized in vitro, there remain many questions on how these cells are generated and their relative importance in host defense. Open questions remain about the molecular mechanisms regulating their enhanced functionality that warrant studies of their mrna, micrornas, protein, phosphorylation, and epigenetic profiles, compared to naïvenkcells.therelationshipofcimltomemorynk cells that arise after MCMV infection requires clarification. The identification of a specific cell surface marker or group of markers that clearly distinguish CIML from naïve NK cells will facilitate their study in healthy and diseased individuals. How are CIML NK cells generated in vivo? We hypothesize that accessory cells (e.g., dendritic cells), which are equipped to produce IL-12, IL-15, and IL-18 and interact with NK cells, represent a physiologic CIML NK cell inducer population. While their ability to control transferred tumor cells is provocative, it remains untested whether CIML NK cells are a vital component of the normal host defense against infections or not. Importantly, while these remain fundamental questions about their biology, our current knowledge of CIML NK cells allows for their rapid translation to the clinic as immunotherapy effectors against cancer. 13. CIML NK Cell Translation NK cells have increasingly been recognized as important contributors to the graft-versus-leukemia effect following allogeneic HSCT, where MHC haploidentical NK cells were identified as alloreactive to myeloid leukemia blasts that lacked the corresponding HLA ligand for at least one KIR receptor and were associated with protection against relapse [152]. Additional potential benefits of NK cells in the context of HSCT have been suggested, including limiting graftversus-host disease by elimination of recipient dendritic cells [152]. More recently, NK cell adoptive immunotherapy platforms have been established to facilitate infusion of allogeneic NK cells enriched from a haploidentical donor leukapheresis [ ]. The NK cell translation group at University of Minnesota was the first to demonstrate adoptive NK cell feasibility and identified lymphodepleting fludarabinecyclophosphamide (Flu-Cy) preparative chemotherapy as a critical component of NK cell expansion, in part through induction of endogenous IL-15. As part of this approach, enriched CD3 CD19 PBMC that contain approximately 50% NK cells (and very few T cells) are activated overnight with high dose IL-2, washed, and infused into the patient. These NK cells are supported by intermediate dose rhil- 2 for approximately 2 weeks following transfer. Using this approach, patients with relapsed or refractory AML obtained complete remissions that correlated with NK cell expansion in vivo, strongly suggesting an NK cell versus leukemia effect; however these remissions were achieved in a minority of patients and did not appear durable. More recently, feasibility of this approach has been shown in other types of cancer, for example, lymphoma patients in combination with the anti-cd20 mab rituximab [156]. Alternative approaches

13 12 Scientifica in clinical testing include exposure to priming tumor cell lysatestoactivatenkcellspriortoadoptivetransferinto AMLpatientsincompleteremission[157] (NCT ). Additional small studies have been reported, including those that include selection of KIR-KIR ligand mismatched NK cells [154, 155]. In another report, purified CD56+CD3 NK cells were administered to pediatric patients with favorable or intermediate risk AML in complete remission (not candidates for immediate allogeneic HSCT), without IL-2 exposure in vitro, but with rhil-2 administered after transfer [155]. While none of the 10 patients who received NK cell infusions in that pilot study relapsed, direct demonstration of the NK cell antileukemia response will require treatment of patients with active disease, or a large randomized study demonstrating improvements in progression free survival. Further, several groups have also infused mature activated NK cells early after allogeneic HSCT to provide NK cell antitumor and anti- GVHD effects [ ]. Several groups have developed ex vivo expansion protocols to help expand and activate NK cells prior to their adoptive transfer into patients [134, ]. One such approach involves genetically modified K562 based feeder cell system with membrane bound IL-21 (mbil- 21) leading to a remarkable NK cell expansion (more than 30-thousand-fold by day 21) and without loss of telomere length reported in some previous studies [134]. While such cells are uniformly highly functional in vitro, clinical trials in patients are required to define the persistence and activity in vivo. Collectively, while all of these varying approaches demonstrate some degree of safety and feasibility, clinical studies are required to definitively show that allogeneic NK cells provide clinical benefit to cancer patients, many of which are in development or ongoing. We hypothesized that human CIML NK cells, based on their enhanced persistence and function against leukemia, will safely provide improved results following adoptive transfer, compared to IL-2-activated or naïve NK cells. To test this hypothesis, we are performing first in human phase 1 study of HLA haploidentical CIML NK cells in patients with relapsed or refractory AML (NCT ), building upon established NK cell adoptive immunotherapy platforms and Flu-Cy conditioning. Haploidentical donors will undergo leukapheresis, followed by selection of CD56+CD3 NK cells, preactivation with IL-12, IL-15, and IL-18 overnight in a GMP facility, extensive washing to remove cytokines, andinfusionintothepatient.further,basedontheclear preclinical data demonstrating a high affinity IL-2Rαβγ on CIML NK cells, low dose ( IU [92]) rhil-2 will be administered for two weeks to support CIML NK cell expansion and functionality. Initial doses will be 5 10-fold lower than typical NK cell infusions to assess for safety of this highly activated NK cell product, followed by dose escalation if well tolerated. While the primary objective of this phase 1 study is safety of the NK cell product, leukemia clearance and responses will be assessed providing some ability to discern NK cell antitumor responses in these patients, especially at the maximal tolerated dose. Further, correlative studies will evaluate key aspects of CIML NK cell biology, providing a unique view of human CIML NK cell biology in vivo. 14. Conclusions NK cell immunotherapy has undergone a renaissance over the past decade, with enticing evidence of graft-versusleukemia/lymphoma effect in the setting of HSCT and evidence of leukemia clearance after adoptive transfer. To date, IL-2 remains the main cytokine utilized in these approaches for in vitro activation and postinfusion maintenance, although rhil-15 and IL-15 mimics are now in clinical testing, and both commercial and NCI/NIH-based productions appear to be expanding. IL-15 has the benefit of activating NK cells without augmenting regulatory T cell function. IL-21 remains highly interesting as an NK cell activator in vivo, especially in combination with approaches that facilitate NK cell recognition such as monoclonal antibodies, and continues in phase 2 clinical development for multiple malignancies. An ex vivo approach to utilize K562 leukemia targets with membrane bound IL-21 also appears promising. While rhil-12 as injected cytokine therapy appeared promising based on preclinical data and early phase clinical studies, rhil-12 is primarily being evaluated in combination with monoclonal antibodies in solid tumors, anticytokine antibodies do occur, and access for clinical trials is limited. It seems likely that continued noncommercial development will be required to further pursue these cytokines as anticancer drugs administered to patients. rhil-18 appeared well tolerated, but a lack of responses has likely stalled clinical development as a single agent. New approaches to harness the potential of cytokine activation include highly translatable and abbreviated ex vivo use, exemplified by our own approach with CIML NK cells. The use of cytokines to function-enable and support NK cells for immunotherapy will require combinatorial approaches that also limit NK cell functionality (anti-kir/anti-pd1 monoclonal antibodies, Treg depletion) and enhance tumor cell recognition (monoclonal antibodies, bi/trispecific targeting reagents, chimeric antigen receptors). Conflict of Interests The authors declare that there is no conflict of interests regarding the publication of this paper. Acknowledgments This work was supported by a Washington University Leukemia SPORE developmental research project (RR, TAF), K08HL (TAF), R01AI (TAF), the Gabrielle s Angel Foundation for Cancer Research, and the V Foundation for Cancer Research (TAF). The authors thank Stephanie E. Schneider for editorial review of the paper. References [1] R. Herberman, M. Nunn, and D. Lavrin, Natural cytotoxic reactivity of mouse lymphoid cells against syngeneic and allogeneic tumors. I. Distribution of reactivity and specificity, International Journal of Cancer,vol.16,no.2,pp ,1975. [2] R. Kiessling, E. Klein, and H. Wigzell, Natural killer cells in the mouse. I. Cytotoxic cells with specificity for mouse

14 Scientifica 13 Moloney leukemia cells. Specificity and distribution according to genotype, European Journal of Immunology, vol. 5, no. 2,pp , [3] M. A. Caligiuri, Human natural killer cells, Blood,vol.112,no. 3, pp , [4] T. Walzer, M. Bléry, J. Chaix et al., Identification, activation, and selective in vivo ablation of mouse NK cells via NKp46, Proceedings of the National Academy of Sciences of the United States of America,vol.104,no.9,pp ,2007. [5]E.Narni-Mancinelli,J.Chaix,A.Fenisetal., Fatemapping analysis of lymphoid cells expressing the NKp46 cell surface receptor, Proceedings of the National Academy of Sciences of the United States of America,vol.108,no.45,pp ,2011. [6] E. Vivier, E. Tomasello, M. Baratin, T. Walzer, and S. Ugolini, Functions of natural killer cells, Nature Immunology, vol.9, no. 5, pp , [7] C. Grégoire,L.Chasson,C.Lucietal., Thetraffickingofnatural killer cells, Immunological Reviews,vol.220,no.1,pp , [8] H. Spits and J. P. di Santo, The expanding family of innate lymphoid cells: regulators and effectors of immunity and tissue remodeling, Nature Immunology, vol. 12,no. 1, pp.21 27, [9] J. S. Orange and Z. K. Ballas, Natural killer cells in human health and disease, Clinical Immunology, vol. 118, no. 1, pp. 1 10, [10] T.W.Kuijpers,P.A.Baars,C.Dantin,M.vandenBurg,R.A.W. vanlier,ande.roosnek, HumanNKcellscancontrolCMV infection in the absence of T cells, Blood,vol.112,no.3,pp , [11] K. Imai, S. Matsuyama, S. Miyake, K. Suga, and K. Nakachi, Natural cytotoxic activity of peripheral-blood lymphocytes and cancer incidence: an 11-year follow-up study of a general population, The Lancet,vol.356,no.9244,pp , [12] K. L. Alderson and P. M. Sondel, Clinical cancer therapy by NK cells via antibody-dependent cell-mediated cytotoxicity, Journal of Biomedicine and Biotechnology, vol. 2011, Article ID , 7 pages, [13] W. J. Murphy, P. Parham, and J. S. Miller, NK cells from bench to clinic, Biology of Blood and Marrow Transplantation, vol. 18, no. 1, supplement, pp. S2 S7, [14] A. Velardi, L. Ruggeri, A. Mancusi, F. Aversa, and F. T. Christiansen, Natural killer cell allorecognition of missing self in allogeneic hematopoietic transplantation: a tool for immunotherapy of leukemia, Current Opinion in Immunology, vol. 21, no. 5, pp , [15] H.-G. Ljunggren and K.-J. Malmberg, Prospects for the use of NK cells in immunotherapy of human cancer, Nature Reviews Immunology,vol.7,no.5,pp ,2007. [16] A. G. Freud and M. A. Caligiuri, Human natural killer cell development, Immunological Reviews,vol.214,no.1,pp.56 72, [17] F. Colucci, M. A. Caligiuri, and J. P. di Santo, What does it take to make a natural killer? Nature Reviews Immunology, vol.3, no. 5, pp , [18] A. G. Freud, A. Yokohama, B. Becknell et al., Evidence for discrete stages of human natural killer cell differentiation in vivo, TheJournalofExperimentalMedicine,vol.203,no.4,pp , [19] A. G. Freud, B. Becknell, S. Roychowdhury et al., A human CD34 + subset resides in lymph nodes and differentiates into CD56 bright natural killer cells, Immunity,vol.22,no.3,pp , [20] J. Miller, K. Alley, and P. McGlave, Differentiation of natural killer (NK) cells from human primitive marrow progenitors in a stroma-based long-term culture system: identification of a CD NK progenitor, Blood, vol.83,no.9,pp , [21] S. Kim, J. Poursine-Laurent, S. M. Truscott et al., Licensing of natural killer cells by host major histocompatibility complex class I molecules, Nature,vol.436,no.7051,pp ,2005. [22]N.D.Huntington,C.A.J.Vosshenrich,andJ.P.diSanto, Developmental pathways that generate natural-killer-cell diversity in mice and humans, Nature Reviews Immunology, vol. 7, no. 9, pp , [23] J. M. Elliott and W. M. Yokoyama, Unifying concepts of MHCdependent natural killer cell education, Trends in Immunology, vol.32,no.8,pp ,2011. [24] N. T. Joncker and D. H. Raulet, Regulation of NK cell responsiveness to achieve self-tolerance and maximal responses to diseased target cells, Immunological Reviews,vol.224,no.1, pp , [25] M. T. Orr and L. L. Lanier, Natural killer cell education and tolerance, Cell, vol. 142, no.6, pp , [26] L. L. Lanier, NK cell recognition, Annual Review of Immunology,vol.23,pp ,2005. [27] S. Cooley, F. Xiao, M. Pitt et al., A subpopulation of human peripheral blood NK cells that lacks inhibitory receptors for self-mhc is developmentally immature, Blood, vol.110,no.2, pp , [28] T. A. Fehniger and M. A. Caligiuri, Interleukin 15: biology and relevance to human disease, Blood, vol. 97, no. 1, pp , [29] T. A. Waldmann, The biology of interleukin-2 and interleukin- 15:implications for cancer therapy and vaccine design, Nature Reviews Immunology,vol.6,no.8,pp ,2006. [30] T. A. Fehniger, S. F. Cai, X. Cao et al., Acquisition of murine NK cell cytotoxicity requires the translation of a pre-existing pool of granzyme B and perforin mrnas, Immunity,vol.26,no.6,pp , [31] M. Lucas, W. Schachterle, K. Oberle, P. Aichele, and A. Diefenbach, Dendritic cells prime natural killer cells by transpresenting interleukin 15, Immunity, vol. 26, no. 4, pp , [32] S.-H. Lee, T. Miyagi, and C. A. Biron, Keeping NK cells in highly regulated antiviral warfare, Trends in Immunology, vol. 28,no.6,pp ,2007. [33] N. D. Huntington, H. Tabarias, K. Fairfax et al., NK cell maturation and peripheral homeostasis is associated with KLRG1 up-regulation, The Journal of Immunology, vol.178,no.8,pp , [34] K. C. Newman and E. M. Riley, Whatever turns you on: accessory-cell-dependent activation of NK cells by pathogens, Nature Reviews Immunology,vol.7,no.4,pp ,2007. [35] L. L. Lanier, Up on the tightrope: natural killer cell activation and inhibition, Nature Immunology, vol.9,no.5,pp , [36] H. A. Jonsson and W. M. Yokoyama, Natural killer cell tolerance licensing and other mechanisms, Advances in Immunology,vol.101,no.8,pp.27 79,2009. [37] I. Tassi, J. Klesney-Tait, and M. Colonna, Dissecting natural killer cell activation pathways through analysis of genetic mutations in human and mouse, Immunological Reviews, vol. 214, no. 1, pp , 2006.

15 14 Scientifica [38] J. M. Venstrom, G. Pittari, T. A. Gooley et al., HLA-Cdependent prevention of leukemia relapse by donor activating KIR2DS1, The New England Journal of Medicine, vol. 367, no. 9, pp ,2012. [39] Y. T. Bryceson and E. O. Long, Line of attack: NK cell specificity and integration of signals, Current Opinion in Immunology,vol. 20, no. 3, pp , [40] L. L. Lanier, Natural killer cell receptor signaling, Current Opinion in Immunology,vol.15,no.3,pp ,2003. [41] S. C. Ganal, S. L. Sanos, C. Kallfass et al., Priming of natural killer cells by nonmucosal mononuclear phagocytes requires instructive signals from commensal microbiota, Immunity,vol. 37, no. 1, pp , [42] J. Chaix, M. S. Tessmer, K. Hoebe et al., Cutting edge: priming of NK cells by IL-18, TheJournalofImmunology,vol.181,no.3, pp ,2008. [43]D.W.White,C.R.Keppel,S.E.Schneideretal., Latent herpesvirus infection arms NK cells, Blood, vol. 115, no. 22, pp , [44] S. Lopez-Verges, J. M. Milush, B. S. Schwartz et al., Expansion of a unique CD57 + NKG2C hi natural killer cell subset during acute human cytomegalovirus infection, Proceedings of the National Academy of Sciences of the United States of America, vol. 108, no. 36, pp , [45]B.Foley,S.Cooley,M.R.Vernerisetal., Cytomegalovirus reactivation after allogeneic transplantation promotes a lasting increase in educated NKG2C + natural killer cells with potent function, Blood,vol.119,no.11,pp ,2012. [46] N. K. Björkström, T.Lindgren, M. Stoltz et al., Rapid expansion and long-term persistence of elevated NK cell numbers in humansinfectedwithhantavirus, TheJournalofExperimental Medicine,vol.208,no.1,pp.13 21,2011. [47] J. S. Orange, Formation and function of the lytic NK-cell immunological synapse, Nature Reviews Immunology, vol.8, no. 9, pp , [48] A. Nagler, L. L. Lanier, S. Cwirla, and J. H. Phillips, Comparative studies of human FcRIII-positive and negative natural killer cells, The Journal of Immunology, vol. 143, no. 10, pp , [49] M. A. Cooper, T. A. Fehniger, S. C. Turner et al., Human natural killer cells: a unique innate immunoregulatory role for the CD56 bright subset, Blood,vol.97,no.10,pp ,2001. [50] T. A. Fehniger, M. A. Cooper, G. J. Nuovo et al., CD56 bright natural killer cells are present in human lymph nodes and are activated by T cell-derived IL-2: a potential new link between adaptive and innate immunity, Blood,vol.101,no.8,pp , [51] M. A. Cooper, J. Punt, T. A. Fehniger, J. Owen, and M. A. Caligiuri, The biology of human natural killer-cell subsets, Trends in Immunology,vol.22,no.11,pp ,2001. [52] E. Wilk, K. Kalippke, S. Buyny, R. E. Schmidt, and R. Jacobs, New aspects of NK cell subset identification and inference of NK cells'regulatory capacity by assessing functional and genomic profiles, Immunobiology, vol. 213, no. 3-4, pp , [53] P. Carrega and G. Ferlazzo, Natural killer cell distribution and trafficking in human tissues, Frontiers in Immunology, vol. 3, article 347, [54] T. Strowig, F. Brilot, F. Arrey et al., Tonsilar NK cells restrict B cell transformation by the epstein-barr virus via IFN-γ, PLoS Pathogens,vol.4,no.2,articlee27,2008. [55] A. R. French and W. M. Yokoyama, Natural killer cells and autoimmunity, Arthritis Research and Therapy,vol.6,no.1,pp. 8 14, [56] G. Ferlazzo, D. Thomas, S.-L. Lin et al., The abundant NK cells in human secondary lymphoid tissues require activation to express killer cell Ig-like receptors and become cytolytic, The Journal of Immunology,vol.172,no.3,pp ,2004. [57] A. de Maria, F. Bozzano, C. Cantoni, and L. Moretta, Revisiting human natural killer cell subset function revealed cytolytic CD56 dim CD16 + NK cells as rapid producers of abundant IFN-γ on activation, Proceedings of the National Academy of Sciences of the United States of America,vol.108,no.2,pp ,2011. [58] C. Fauriat, E. O. Long, H.-G. Ljunggren, and Y. T. Bryceson, Regulation of human NK-cell cytokine and chemokine production by target cell recognition, Blood, vol. 115, no. 11, pp , [59] S. Lopez-Vergès, J. M. Milush, S. Pandey et al., CD57 defines a functionally distinct population of mature NK cells in the human CD56 dim CD16 + NK-cell subset, Blood, vol.116,no.19, pp , [60] J. Yu, H. C. Mao, M. Wei et al., CD94 surface density identifies a functional intermediary between the CD56 dim and CD56 bright human NK-cell subsets, Blood, vol. 115, no. 2, pp , [61] N. K. Björkström, P. Riese, F. Heuts et al., Expression patterns of NKG2A, KIR, and CD57 define a process of CD56 dim NK-cell differentiation uncoupled from NK-cell education, Blood, vol. 116,no.19,pp ,2010. [62] R. Romee, J. Leong, S. Schneider, R. Sullivan, and T. Fehniger, IL-15 primes a highly potent anti-leukemia response by CD56 bright NK cells, Blood, vol.122,no.21,articleida2283, [63] A. Chan, D.-L. Hong, A. Atzberger et al., CD56 dim human NK cells differentiate into CD56 bright cells: role of contact with peripheral fibroblasts, The Journal of Immunology, vol. 179, no. 1,pp.89 94,2013. [64] N. D. Huntington, N. Legrand, N. L. Alves et al., IL-15 trans-presentation promotes human NK cell development and differentiation in vivo, The Journal of Experimental Medicine, vol. 206, no. 1, pp , [65] S. A. Dezell, Y.-O. Ahn, J. Spanholtz et al., Natural killer cell differentiation from hematopoietic stem cells: a comparative analysis of heparin- and stromal cell-supported methods, Biology of Blood and Marrow Transplantation,vol.18,no.4,pp , [66] E. M. Mace, A. P. Hsu, L. Monaco-Shawver et al., Mutations in GATA2 cause human NK cell deficiency with specific loss of the CD56 bright subset, Blood,vol.121,no.14,pp ,2013. [67] M. J. Robertson and J. Ritz, Biology and clinical relevance of human natural killer cells, Blood,vol.76,no.12,pp , [68] J. H. Russell and T. J. Ley, Lymphocyte-mediated cytotoxicity, Annual Review of Immunology,vol.20,no.6,pp ,2002. [69] H. Braumüller, T. Wieder, E. Brenner et al., T-helper-1-cell cytokines drive cancer into senescence, Nature, vol. 494, no. 7437, pp , [70] T. A. Fehniger, M. H. Shah, M. J. Turner et al., Differential cytokine and chemokine gene expression by human NK cells following activation with IL-18 or IL-15 in combination with IL- 12: implications for the innate immune response, The Journal of Immunology,vol.162,no.8,pp ,1999.

16 Scientifica 15 [71] M. T. Orr, W. J. Murphy, and L. L. Lanier, Unlicensed natural killer cells dominate the response to cytomegalovirus infection, Nature Immunology,vol.11,no.4,pp ,2010. [72] L.Yang,Y.Pang,andH.L.Moses, TGF-β and immune cells: an important regulatory axis in the tumor microenvironment and progression, Trends in Immunology,vol.31,no.6,pp , [73]E.B.Wilson,J.J.El-Jawhari,A.L.Neilsonetal., Human tumour immune evasion via TGF-β blocks NK cell activation but not survival allowing therapeutic restoration of anti-tumour activity, PLoS ONE,vol.6,no.9,ArticleIDe22842,2011. [74] T. A. Fehniger, M. A. Cooper, and M. A. Caligiuri, Interleukin- 2 and interleukin-15: immunotherapy for cancer, Cytokine & Growth Factor Reviews,vol.13,no.2,pp ,2002. [75] B. Becknell and M. A. Caligiuri, Interleukin-2, interleukin- 15, and their roles in human natural killer cells, Advances in Immunology,vol.86,pp ,2005. [76] M. A. Caligiuri, A. Zmuidzinas, T. J. Manley, H. Levine, K. A. Smith, and J. Ritz, Functional consequences of interleukin 2 receptor expression on resting human lymphocytes. Identification of a novel natural killer cell subset with high affinity receptors, TheJournalofExperimentalMedicine, vol. 171, no. 5, pp , [77] A. Nagler, L. Lanier, and J. Phillips, Constitutive expression of high affinity interleukin 2 receptors on human CD16 natural killer cells in vivo, TheJournalofExperimentalMedicine,vol. 171, no. 5, pp , [78] J. Leong, J. Chase, R. Romee, S. Schneider, R. Sullivan, and T. Fehniger, Cytokine activation induces CD25 expression and a functional high-affinity IL-2 receptor on CD56 dim human NK Cells, The Journal of Immunology,vol.190,2014. [79]A.-H.Pillet,F.Bugault,J.Thèze,L.A.Chakrabarti,andT. Rose, AprogrammedswitchfromIL-15-toIL-2-dependent activation in human NK cells, The Journal of Immunology,vol. 182, no. 10, pp , [80] A.-H. Pillet, J. Thèze, and T. Rose, Interleukin (IL)-2 and IL- 15 have different effects on human natural killer lymphocytes, Human Immunology, vol. 72, no. 11, pp , [81] S. Rosenberg, Interleukin-2 and the development of immunotherapy for the treatment of patients with cancer, Cancer Journal from Scientific American, vol.6,supplement1, pp.s2 S7,2000. [82] B. Y. H. Wang and K. A. Smith, The IL-2 receptor: functional consequences of its bimolecular structure, The Journal of Experimental Medicine, vol. 166, no. 4, pp , [83] M. A. Caligiuri, C. Murray, R. Soiffer et al., Extended continuous infusion low-dose recombinant interleukin-2 in advanced cancer: prolonged immunomodulation without significant toxicity, JournalofClinicalOncology, vol. 9, no. 12, pp , [84] Z. P. Bernstein, M. M. Porter, M. Gould et al., Prolonged administration of low-dose interleukin-2 in human immunodeficiency virus-associated malignancy results in selective expansion of innate immune effectors without significant clinical toxicity, Blood,vol.86,no.9,pp , [85] M. A. Caligiuri, C. Murray, M. Robertson et al., Selective modulation of human natural killer cells in vivo after prolonged infusion of low dose recombinant interleukin 2, The Journal of Clinical Investigation,vol.91,no.1,pp ,1993. [86] N. J. Meropol, G. M. Barresi, T. Fehniger, J. Hitt, M. Franklin, and M. Caligiuri, Evaluation of natural killer cell expansion and activation in vivo with daily subcutaneous low-dose interleukin-2 plus periodic intermediate-dose pulsing, Cancer Immunology Immunotherapy, vol. 46, no. 6, pp , [87] V. P. Khatri, T. Fehniger, R. A. Baiocchi et al., Ultra low dose interleukin-2 therapy promotes a type 1 cytokine profile in vivo in patients with AIDS and AIDS-associated malignancies, The JournalofClinicalInvestigation,vol.101,no.6,pp , [88] T. Fehniger, E. Bluman, M. Porter et al., Potential mechanisms of human natural killer cell expansion in vivo during low-dose IL-2 therapy, The Journal of Clinical Investigation, vol.106,no. 1,pp ,2000. [89] P. Sondel and J. Hank, Combination therapy with interleukin- 2 and antitumor monoclonal antibodies, Cancer Journal from Scientific American,vol.3,supplement1,pp.S121 S127,1997. [90] T. R. Malek and I. Castro, Interleukin-2 receptor signaling: at the interface between tolerance and immunity, Immunity, vol. 33,no.2,pp ,2010. [91] M. Ahmadzadeh and S. A. Rosenberg, IL-2 administration increases CD4 + CD25 hi Foxp3 + regulatory T cells in cancer patients, Blood,vol.107,no.6,pp ,2006. [92] J. Koreth, K. Matsuoka, H. Kim et al., Interleukin-2 and regulatory T cells in graft-versus-host disease, The New England Journal of Medicine,vol.365,no.22,pp ,2011. [93] E. Zorn, E. A. Nelson, M. Mohseni et al., IL-2 regulates FOXP3 expression in human CD4 + CD25 + regulatory T cells through a STAT-dependent mechanism and induces the expansion of these cells in vivo, Blood, vol. 108, no. 5,pp , [94] A. Ma, R. Koka, and P. Burkett, Diverse functions of IL-2, IL-15, and IL-7 in lymphoid homeostasis, Annual Review of Immunology,vol.24,pp ,2006. [95] C. Berger, M. Berger, R. C. Hackman et al., Safety and immunologic effects of IL-15 administration in nonhuman primates, Blood,vol.114,no. 12,pp , [96] S. Cooley, M. R. Verneris, J. Curtsinger et al., Recombinant human IL-15 promotes in vivo expansion of adoptively transferred NK cells in a first-in-human phase I dose escalation study in patients with AML, Blood,vol.120,no.21,articleA894,2012. [97] M. P. Rubinstein, M. Kovar, J. F. Purton et al., Converting IL-15 to a superagonist by binding to soluble IL-15Rα, Proceedings of the National Academy of Sciences of the United States of America, vol. 103, no. 24, pp , [98] T. A. Stoklasek, K. S. Schluns, and L. Lefranc, Combined IL- 15/IL-15Rα immunotherapy maximizes IL-15 activity in vivo, TheJournalofImmunology,vol.177,no.9,pp ,2006. [99] X. Zhu, W. D. Marcus, W. Xu et al., Novel human interleukin-15 agonists, The Journal of Immunology, vol.183,no.6,pp , [100] K. Han, X. Zhu, B. Liu et al., IL-15: IL-15 receptor alpha superagonist complex: high-level co-expression in recombinant mammalian cells, purification and characterization, Cytokine, vol.56,no.3,pp ,2011. [101] M. Kobayashi, L. Fitz, M. Ryan et al., Identification and purification of natural killer cell stimulatory factor (NKSF), a cytokine with multiple biologic effects on human lymphocytes, The Journal of Experimental Medicine, vol.170,no.3,pp , [102] M. K. Gately, L. M. Renzetti, J. Magram et al., The interleukin- 12/interleukin-12-receptor system: role in normal and pathologic immune responses, Annual Review of Immunology, vol. 12, pp , 1998.

17 16 Scientifica [103]M.J.Robertson,R.J.Soiffer,S.F.Wolfetal., Responseof human natural killer (NK) cells to NK cell stimulatory factor (NKSF): cytolytic activity and proliferation of NK cells are differentially regulated by NKSF, The Journal of Experimental Medicine,vol.175,no.3,pp ,1992. [104] K. S. Wang, D. A. Frank, and J. Ritz, Interleukin-2 enhances the response of natural killer cells to interleukin-12 through upregulation of the interleukin-12 receptor and STAT4, Blood, vol. 95,no.10,pp ,2000. [105]R.Parihar,J.Dierksheide,Y.Hu,andW.E.Carson, IL-12 enhances the natural killer cell cytokine response to Ab-coated tumor cells, The Journal of Clinical Investigation,vol.110,no.7, pp ,2002. [106] B. M. J. Brunda, L. Luistro, R. R. Warrier et al., Antitumor and antimetastatic activity of interleukin 12 against murine tumors, TheJournalofExperimentalMedicine,vol.178,no.4,pp , [107] C.Nastala,H.D.Edington,T.C.McKinneyetal., Recombinant IL-12 administration induces tumor regression in association with IFN-γ production, TheJournalofImmunology, vol. 153, no. 4, pp , [108] M. B. Atkins, M. J. Robertson, M. Gordon et al., Phase I evaluation of intravenous recombinant human interleukin 12 in patients with advanced malignancies, Clinical Cancer Research, vol. 3, no. 3, pp , [109] J.P.Leonard,M.L.Sherman,G.L.Fisheretal., Effectsofsingledose interleukin-12 exposure on interleukin-12 associated toxicity and interferon-γ production, Blood, vol. 90, no. 7, pp , [110] E. Bajetta, M. del Vecchio, R. Mortarini et al., Pilot study of subcutaneous recombinant human interleukin 12 in metastatic melanoma, Clinical Cancer Research, vol. 4, no. 1, pp , [111] M. J. Robertson, C. Cameron, M. B. Atkins et al., Immunological effects of interleukin 12 administered by bolus intravenous injection to patients with cancer, Clinical Cancer Research,vol. 5,no.1,pp.9 16,1999. [112]R.J.Motzer,A.Rakhit,J.A.Thompsonetal., Randomized multicenter phase II trial of subcutaneous recombinant human interleukin-12 versus interferon-α2aforpatientswithadvanced renal cell carcinoma, Journal of Interferon & Cytokine Research, vol. 21, no. 4, pp , [113] J. M. Roda, T. Joshi, J. P. Butchar et al., The activation of natural killer cell effector functions by cetuximab-coated, epidermal growth factor receptor positive tumor cells is enhanced by cytokines, Clinical Cancer Research, vol.13,no.21,pp , [114] S. V. Kondadasula, J. M. Roda, R. Parihar et al., Colocalization of the IL-12 receptor and FcγRIIIa to natural killer cell lipid rafts leads to activation of ERK and enhanced production of interferon-γ, Blood, vol. 111,no. 8, pp , [115] R. Parihar, P. Nadella, A. Lewis et al., A phase I study of interleukin 12 with trastuzumab in patients with human epidermal growth factor receptor-2-overexpressing malignancies: analysis of sustained interferon γ production in a subset of patients, Clinical Cancer Research,vol.10,no.15,pp ,2004. [116] C. F. Eisenbeis, G. B. Lesinski, M. Anghelina et al., Phase I study of the sequential combination of interleukin-12 and iterferon alfa-2b in advanced cancer: evidence for modulation of interferon signaling pathways by interleukin-12, Journal of Clinical Oncology, vol. 23, no. 34, pp , [117]S.M.Ansell,S.M.Geyer,M.J.Maureretal., Randomized phase II study of interleukin-12 in combination with rituximab in previously treated non-hodgkin's lymphoma patients, Clinical Cancer Research, vol. 12, no. 20, pp , [118] C. A. Dinarello, D. Novick, S. Kim, and G. Kaplanski, Interleukin-18 and IL-18 binding protein, Frontiers in Immunology,vol.4,article289,2013. [119] U. Kalina, D. Kauschat, N. Koyama et al., IL-18 activates STAT3 in the natural killer cell line 92, augments cytotoxic activity, and mediates IFN-γ production by the stress kinase p38 and by the extracellular regulated kinases p44 erk-1 and p42 erk-2, The Journal of Immunology,vol.165,no.3,pp ,2000. [120]A.R.French,E.B.Holroyd,L.Yang,S.Kim,andW.M. Yokoyama, IL-18 acts synergistically with IL-15 in stimulating natural killer cell proliferation, Cytokine, vol.35,no.5-6,pp , [121]T.Yoshimoto,K.Takeda,T.Tanakaetal., IL-12up-regulates IL-18 receptor expression on T cells, Th1 cells, and B cells: synergism with IL-18 for IFN-γ production, The Journal of Immunology, vol. 161, no. 7, pp , [122] K. Takeda, H. Tsutsui, T. Yoshimoto et al., Defective NK cell activity and Th1 response in IL-18-deficient mice, Immunity, vol. 8, no. 3, pp , [123] M. J. Robertson, J. W. Mier, T. Logan et al., Clinical and biological effects of recombinant human interleukin-18 administered by intravenous infusion to patients with advanced cancer, Clinical Cancer Research, vol.12,no.14,part1,pp , [124] M.J.Robertson,J.Kline,H.Struemperetal., Adose-escalation study of recombinant human interleukin-18 in combination with rituximab in patients with non-hodgkin lymphoma, Journal of Immunotherapy,vol.36,no.6,pp ,2013. [125]M.J.Robertson,J.M.Kirkwood,T.F.Loganetal., Adoseescalation study of recombinant human interleukin-18 using two different schedules ofadministration in patients with cancer, Clinical Cancer Research, vol.14,no.11,pp , [126]K.Ozaki,K.Kikly,D.Michalovich,P.R.Young,andW.J. Leonard, Cloning of a type I cytokine receptor most related to the IL-2 receptor β chain, Proceedings of the National Academy of Sciences of the United States of America, vol.97,no.21,pp , [127] R. Zeng, R. Spolski, E. Casas, W. Zhu, D. E. Levy, and W. J. Leonard, The molecular basis of IL-21-mediated proliferation, Blood,vol.109,no.10,pp ,2007. [128] H. Jin, R. Carrio, A. Yu, and T. R. Malek, Distinct activation signals determine whether IL-21 induces B cell costimulation, growth arrest, or Bim-dependent apoptosis, The Journal of Immunology,vol.173,no.1,pp ,2004. [129]B.R.Barker,J.G.Parvani,D.Meyer,A.S.Hey,K.Skak,and N. L. Letvin, IL-21 induces apoptosis of antigen-specific CD8 + Tlymphocytes, The Journal of Immunology,vol.179,no.6,pp , [130] J. Pène,J.-F.Gauchat,S.Lécartetal., Cuttingedge:IL-21isa switch factor for the production of IgG 1 and IgG3 by human B cells, The Journal of Immunology,vol.172,no.9,pp , [131] T. Korn, E. Bettelli, W. Gao et al., IL-21 initiates an alternative pathway to induce proinflammatory T H 17 cells, Nature, vol. 448,no.7152,pp ,2007.

18 Scientifica 17 [132] J. Brady, Y. Hayakawa, M. J. Smyth, and S. L. Nutt, IL-21 induces the functional maturation of murine NK cells, The Journal of Immunology, vol. 172, no. 4, pp , [133] R. Takaki, Y. Hayakawa, A. Nelson et al., IL-21 enhances tumor rejection through a NKG2D-dependent mechanism, The Journal of Immunology,vol.175,no.4,pp ,2005. [134] C. J. Denman, V. V. Senyukov, S. S. Somanchi et al., Membranebound IL-21 promotes sustained ex vivo proliferation of human natural killer cells, PLoS ONE, vol.7,no.1,articleide30264, [135] I. D. Davis, B. K. Skrumsager, J. Cebon et al., An open-label, two-arm, phase I trial of recombinant human interleukin-21 in patients with metastatic melanoma, Clinical Cancer Research, vol.13,no.12,pp ,2007. [136] I. D. Davis, B. Brady, R. F. Kefford et al., Clinical and biological efficacy of recombinant human interleukin-21 in patients with stageivmalignantmelanomawithoutpriortreatment:aphase lla trial, Clinical Cancer Research, vol.15,no.6,pp , [137] T.M.Petrella,R.Tozer,K.Belangeretal., Interleukin-21has activity in patients with metastatic melanoma: a phase II study, Journal of Clinical Oncology,vol.30,no.27,pp ,2012. [138] N. Steele, A. Anthony, M. Saunders et al., A phase 1 trial of recombinant human IL-21 in combination with cetuximab in patients with metastatic colorectal cancer, British Journal of Cancer,vol.106,no.5,pp ,2012. [139] A. Gowda, J. Roda, S.-R. A. Hussain et al., IL-21 mediates apoptosis through up-regulation of the BH3 family member BIM and enhances both direct and antibody-dependent cellular cytotoxicity in primary chronic lymphocytic leukemia cells in vitro, Blood,vol. 111,no.9, pp , [140] J. C. Sun, J. N. Beilke, and L. L. Lanier, Adaptive immune features of natural killer cells, Nature, vol. 457, no. 7229, pp , [141]J.G.O'Leary,M.Goodarzi,D.L.Drayton,andU.H.von Andrian, T cell- and B cell-independent adaptive immunity mediated by natural killer cells, Nature Immunology,vol.7,no. 5, pp , [142] M.A.Cooper,J.M.Elliott,P.A.Keyel,L.Yang,J.A.Carrero, and W. M. Yokoyama, Cytokine-induced memory-like natural killer cells, Proceedings of the National Academy of Sciences of the United States of America,vol.106,no.6,pp ,2009. [143] G. Min-Oo, Y. Kamimura, D. W. Hendricks, T. Nabekura, and L. L. Lanier, Natural killer cells: walking three paths down memory lane, Trends in Immunology, vol. 34, no. 6, pp , [144] M. A. Cooper, M. Colonna, and W. M. Yokoyama, Hidden talents of natural killers: NK cells in innate and adaptive immunity, The EMBO Reports, vol. 10, no. 10, pp , [145] M. Keppel, L. Yang, and M. Cooper, Murine NK cell intrinsic cytokine-induced memory-like responses are maintained following homeostatic proliferation, The Journal of Immunology, vol. 190, no. 9, pp , [146] J. C. Sun, J. N. Beilke, N. A. Bezman, and L. L. Lanier, Homeostatic proliferation generates long-lived natural killer cells that respond against viral infection, The Journal of Experimental Medicine,vol.208,no.2,pp ,2011. [147] R. Romee, S. E. Schneider, J. W. Leong et al., Cytokine activation induces human memory-like NK cells, Blood, vol. 120, no. 24, pp , [148] Paper: Human Cytokine-Induced Memory-Like (CIML) NK Cells Exhibit Potent Anti-Leukemia Cytotoxicity and Maintain Memory-Like Functionality after Adoptive Transfer Into Immunodeficient NOD-SCID-Gc / (NSG) Mice, [149] J. Ni, M. Miller, A. Stojanovic, N. Garbi, and A. Cerwenka, Sustained effector function of IL-12/15/18-preactivated NK cells against established tumors, The Journal of Experimental Medicine,vol.209,no.13,pp ,2012. [150] J. W. Leong, J. M. Chase, R. Romee et al., Preactivation with IL- 12, IL-15, and IL-18 induces CD25 and a functional high-affinity IL-2 receptor on human cytokine-induced memory-like natural killer cells, Biology of Blood and Marrow Transplantation, vol. 20,no.4,pp ,2014. [151] G. Gasteiger, S. Hemmers, M. A. Firth et al., IL-2-dependent tuning of NK cell sensitivity for target cells is controlled by regulatory T cells, The Journal of Experimental Medicine, vol. 210,no.6,pp ,2013. [152] L. Ruggeri, M. Capanni, E. Urbani et al., Effectiveness of donor natural killer cell aloreactivity in mismatched hematopoietic transplants, Science,vol.295,no.5562,pp ,2002. [153] J. S. Miller, Y. Soignier, A. Panoskaltsis-Mortari et al., Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer, Blood, vol. 105, no. 8, pp , [154] A. Curti, L. Ruggeri, A. D'Addio et al., Successful transfer of alloreactive haploidentical KIR ligand-mismatched natural killer cells after infusion in elderly high risk acute myeloid leukemia patients, Blood,vol.118,no.12,pp ,2011. [155] J. E. Rubnitz, H. Inaba, R. C. Ribeiro et al., NKAML: a pilot study to determine the safety and feasibility of haploidentical natural killer cell transplantation in childhood acute myeloid leukemia, Journal of Clinical Oncology, vol.28,no.6,pp , [156] V. Bachanova, L. J. Burns, D. H. McKenna et al., Allogeneic natural killer cells for refractory lymphoma, Cancer Immunology, Immunotherapy,vol.59,no.11,pp ,2010. [157] J. North, I. Bakhsh, C. Marden et al., Tumor-primed human natural killer cells lyse NK-resistant tumor targets: evidence of a two-stage process in resting NK cell activation, The Journal of Immunology,vol.178,no.1,pp.85 94,2007. [158] U. Koehl, J. Sörensen, R. Esser et al., IL-2 activated NK cell immunotherapy of three children after haploidentical stem cell transplantation, Blood Cells, Molecules, and Diseases, vol. 33, no.3,pp ,2004. [159] J. R. Passweg, A. Tichelli, S. Meyer-Monard et al., Purified donor NK-lymphocyte infusion to consolidate engraftment after haploidentical stem cell transplantation, Leukemia, vol. 18, no. 11, pp , [160] D. A. Rizzieri, R. Storms, D.-F. Chen et al., Natural killer cell-enriched donor lymphocyte infusions from A 3-6/6 HLA matched family member following nonmyeloablative allogeneic stem cell transplantation, Biology of Blood and Marrow Transplantation, vol. 16, no. 8, pp , [161] C. Brehm, S. Huenecke, A. Quaiser et al., IL-2 stimulated but not unstimulated NK cells induce selective disappearance of peripheral blood cells: concomitant results to a phase I/II study, PLoS ONE,vol.6,no.11,ArticleIDe27351,2011. [162] E. Alici, T. Sutlu, B. Björkstrand et al., Autologous antitumor activity by NK cells expanded from myeloma patients using GMP-compliant components, Blood, vol. 111, no. 6, pp , 2008.

19 18 Scientifica [163] T. Sutlu, B. Stellan, M. Gilljam et al., Clinical-grade, large-scale, feeder-free expansion of highly active human natural killer cells for adoptive immunotherapy using an automated bioreactor, Cytotherapy,vol.12,no.8,pp ,2010. [164] W. Lin, C. J. Voskens, X. Zhang et al., Fc-dependent expression of CD137 on human NK cells: insights into agonistic effects of anti-cd137 monoclonal antibodies, Blood, vol. 112, no. 3,pp , [165] R.W.ChildsandM.Berg, Bringingnaturalkillercellstothe clinic: ex vivo manipulation, Hematology: American Society of Hematology Education Program, vol.2013,no.1,pp , [166]D.M.Baume,M.J.Robertson,H.Levine,T.J.Manley,P. W. Schow, and J. Ritz, Differential responses to interleukin 2 define functionally distinct subsets of human natural killer cells, European Journal of Immunology, vol. 22, no. 1, pp. 1 6, 1992.

20 MEDIATORS of INFLAMMATION The Scientific World Journal Gastroenterology Research and Practice Journal of Diabetes Research International Journal of Journal of Endocrinology Immunology Research Disease Markers Submit your manuscripts at BioMed Research International PPAR Research Journal of Obesity Journal of Ophthalmology Evidence-Based Complementary and Alternative Medicine Stem Cells International Journal of Oncology Parkinson s Disease Computational and Mathematical Methods in Medicine AIDS Behavioural Neurology Research and Treatment Oxidative Medicine and Cellular Longevity

Natural Killer Cells: Development, Diversity, and Applications to Human Disease Dr. Michael A. Caligiuri

Natural Killer Cells: Development, Diversity, and Applications to Human Disease Dr. Michael A. Caligiuri Natural Killer Cells: Development, Diversity, November 26, 2008 The Ohio State University Comprehensive Cancer Center The James Cancer Hospital and Solove Research Institute Columbus, Ohio, USA 1 Human

More information

Scott Abrams, Ph.D. Professor of Oncology, x4375 Kuby Immunology SEVENTH EDITION

Scott Abrams, Ph.D. Professor of Oncology, x4375 Kuby Immunology SEVENTH EDITION Scott Abrams, Ph.D. Professor of Oncology, x4375 scott.abrams@roswellpark.org Kuby Immunology SEVENTH EDITION CHAPTER 13 Effector Responses: Cell- and Antibody-Mediated Immunity Copyright 2013 by W. H.

More information

NKTR-255: Accessing The Immunotherapeutic Potential Of IL-15 for NK Cell Therapies

NKTR-255: Accessing The Immunotherapeutic Potential Of IL-15 for NK Cell Therapies NKTR-255: Accessing The Immunotherapeutic Potential Of IL-15 for NK Cell Therapies Saul Kivimäe Senior Scientist, Research Biology Nektar Therapeutics NK Cell-Based Cancer Immunotherapy, September 26-27,

More information

Tumor Immunology. Wirsma Arif Harahap Surgical Oncology Consultant

Tumor Immunology. Wirsma Arif Harahap Surgical Oncology Consultant Tumor Immunology Wirsma Arif Harahap Surgical Oncology Consultant 1) Immune responses that develop to cancer cells 2) Escape of cancer cells 3) Therapies: clinical and experimental Cancer cells can be

More information

Shiv Pillai Ragon Institute, Massachusetts General Hospital Harvard Medical School

Shiv Pillai Ragon Institute, Massachusetts General Hospital Harvard Medical School CTLs, Natural Killers and NKTs 1 Shiv Pillai Ragon Institute, Massachusetts General Hospital Harvard Medical School CTL inducing tumor apoptosis 3 Lecture outline CD8 + Cytotoxic T lymphocytes (CTL) Activation/differentiation

More information

Immune Checkpoints. PD Dr med. Alessandra Curioni-Fontecedro Department of Hematology and Oncology Cancer Center Zurich University Hospital Zurich

Immune Checkpoints. PD Dr med. Alessandra Curioni-Fontecedro Department of Hematology and Oncology Cancer Center Zurich University Hospital Zurich Immune Checkpoints PD Dr med. Alessandra Curioni-Fontecedro Department of Hematology and Oncology Cancer Center Zurich University Hospital Zurich Activation of T cells requires co-stimulation Science 3

More information

NKTR-255: Accessing IL-15 Therapeutic Potential through Robust and Sustained Engagement of Innate and Adaptive Immunity

NKTR-255: Accessing IL-15 Therapeutic Potential through Robust and Sustained Engagement of Innate and Adaptive Immunity NKTR-255: Accessing IL-15 Therapeutic Potential through Robust and Sustained Engagement of Innate and Adaptive Immunity Peiwen Kuo Scientist, Research Biology Nektar Therapeutics August 31 st, 2018 Emerging

More information

T-cell activation T cells migrate to secondary lymphoid tissues where they interact with antigen, antigen-presenting cells, and other lymphocytes:

T-cell activation T cells migrate to secondary lymphoid tissues where they interact with antigen, antigen-presenting cells, and other lymphocytes: Interactions between innate immunity & adaptive immunity What happens to T cells after they leave the thymus? Naïve T cells exit the thymus and enter the bloodstream. If they remain in the bloodstream,

More information

T-cell activation T cells migrate to secondary lymphoid tissues where they interact with antigen, antigen-presenting cells, and other lymphocytes:

T-cell activation T cells migrate to secondary lymphoid tissues where they interact with antigen, antigen-presenting cells, and other lymphocytes: Interactions between innate immunity & adaptive immunity What happens to T cells after they leave the thymus? Naïve T cells exit the thymus and enter the bloodstream. If they remain in the bloodstream,

More information

Chapter 10 (pages ): Differentiation and Functions of CD4+ Effector T Cells Prepared by Kristen Dazy, MD, Scripps Clinic Medical Group

Chapter 10 (pages ): Differentiation and Functions of CD4+ Effector T Cells Prepared by Kristen Dazy, MD, Scripps Clinic Medical Group FIT Board Review Corner September 2015 Welcome to the FIT Board Review Corner, prepared by Andrew Nickels, MD, and Sarah Spriet, DO, senior and junior representatives of ACAAI's Fellows-In-Training (FITs)

More information

ACTIVATION AND EFFECTOR FUNCTIONS OF CELL-MEDIATED IMMUNITY AND NK CELLS. Choompone Sakonwasun, MD (Hons), FRCPT

ACTIVATION AND EFFECTOR FUNCTIONS OF CELL-MEDIATED IMMUNITY AND NK CELLS. Choompone Sakonwasun, MD (Hons), FRCPT ACTIVATION AND EFFECTOR FUNCTIONS OF CELL-MEDIATED IMMUNITY AND NK CELLS Choompone Sakonwasun, MD (Hons), FRCPT Types of Adaptive Immunity Types of T Cell-mediated Immune Reactions CTLs = cytotoxic T lymphocytes

More information

Scott Abrams, Ph.D. Professor of Oncology, x4375 Kuby Immunology SEVENTH EDITION

Scott Abrams, Ph.D. Professor of Oncology, x4375 Kuby Immunology SEVENTH EDITION Scott Abrams, Ph.D. Professor of Oncology, x4375 scott.abrams@roswellpark.org Kuby Immunology SEVENTH EDITION CHAPTER 11 T-Cell Activation, Differentiation, and Memory Copyright 2013 by W. H. Freeman and

More information

Cytokines modulate the functional activities of individual cells and tissues both under normal and pathologic conditions Interleukins,

Cytokines modulate the functional activities of individual cells and tissues both under normal and pathologic conditions Interleukins, Cytokines http://highered.mcgraw-hill.com/sites/0072507470/student_view0/chapter22/animation the_immune_response.html Cytokines modulate the functional activities of individual cells and tissues both under

More information

Adaptive immune responses: T cell-mediated immunity

Adaptive immune responses: T cell-mediated immunity MICR2209 Adaptive immune responses: T cell-mediated immunity Dr Allison Imrie allison.imrie@uwa.edu.au 1 Synopsis: In this lecture we will discuss the T-cell mediated immune response, how it is activated,

More information

Central tolerance. Mechanisms of Immune Tolerance. Regulation of the T cell response

Central tolerance. Mechanisms of Immune Tolerance. Regulation of the T cell response Immunoregulation: A balance between activation and suppression that achieves an efficient immune response without damaging the host. Mechanisms of Immune Tolerance ACTIVATION (immunity) SUPPRESSION (tolerance)

More information

Mechanisms of Immune Tolerance

Mechanisms of Immune Tolerance Immunoregulation: A balance between activation and suppression that achieves an efficient immune response without damaging the host. ACTIVATION (immunity) SUPPRESSION (tolerance) Autoimmunity Immunodeficiency

More information

ACTIVATION OF T LYMPHOCYTES AND CELL MEDIATED IMMUNITY

ACTIVATION OF T LYMPHOCYTES AND CELL MEDIATED IMMUNITY ACTIVATION OF T LYMPHOCYTES AND CELL MEDIATED IMMUNITY The recognition of specific antigen by naïve T cell induces its own activation and effector phases. T helper cells recognize peptide antigens through

More information

General Overview of Immunology. Kimberly S. Schluns, Ph.D. Associate Professor Department of Immunology UT MD Anderson Cancer Center

General Overview of Immunology. Kimberly S. Schluns, Ph.D. Associate Professor Department of Immunology UT MD Anderson Cancer Center General Overview of Immunology Kimberly S. Schluns, Ph.D. Associate Professor Department of Immunology UT MD Anderson Cancer Center Objectives Describe differences between innate and adaptive immune responses

More information

Question 1. Kupffer cells, microglial cells and osteoclasts are all examples of what type of immune system cell?

Question 1. Kupffer cells, microglial cells and osteoclasts are all examples of what type of immune system cell? Abbas Chapter 2: Sarah Spriet February 8, 2015 Question 1. Kupffer cells, microglial cells and osteoclasts are all examples of what type of immune system cell? a. Dendritic cells b. Macrophages c. Monocytes

More information

M.Sc. III Semester Biotechnology End Semester Examination, 2013 Model Answer LBTM: 302 Advanced Immunology

M.Sc. III Semester Biotechnology End Semester Examination, 2013 Model Answer LBTM: 302 Advanced Immunology Code : AS-2246 M.Sc. III Semester Biotechnology End Semester Examination, 2013 Model Answer LBTM: 302 Advanced Immunology A. Select one correct option for each of the following questions:- 2X10=10 1. (b)

More information

Adaptive Immunity. Jeffrey K. Actor, Ph.D. MSB 2.214,

Adaptive Immunity. Jeffrey K. Actor, Ph.D. MSB 2.214, Adaptive Immunity Jeffrey K. Actor, Ph.D. MSB 2.214, 500-5344 Lecture Objectives: Understand role of various molecules including cytokines, chemokines, costimulatory and adhesion molecules in the development

More information

T cells III: Cytotoxic T lymphocytes and natural killer cells

T cells III: Cytotoxic T lymphocytes and natural killer cells T cells III: Cytotoxic T lymphocytes and natural killer cells Margrit Wiesendanger Division of Rheumatology, CUMC September 17, 2008 Killer cells: CD8 + T cells (adaptive) vs. natural killer (innate) Shared

More information

Micro 204. Cytotoxic T Lymphocytes (CTL) Lewis Lanier

Micro 204. Cytotoxic T Lymphocytes (CTL) Lewis Lanier Micro 204 Cytotoxic T Lymphocytes (CTL) Lewis Lanier Lewis.Lanier@ucsf.edu Lymphocyte-mediated Cytotoxicity CD8 + αβ-tcr + T cells CD4 + αβ-tcr + T cells γδ-tcr + T cells Natural Killer cells CD8 + αβ-tcr

More information

Tumor Immunology: A Primer

Tumor Immunology: A Primer Transcript Details This is a transcript of a continuing medical education (CME) activity accessible on the ReachMD network. Additional media formats for the activity and full activity details (including

More information

CYTOKINE RECEPTORS AND SIGNAL TRANSDUCTION

CYTOKINE RECEPTORS AND SIGNAL TRANSDUCTION CYTOKINE RECEPTORS AND SIGNAL TRANSDUCTION What is Cytokine? Secreted popypeptide (protein) involved in cell-to-cell signaling. Acts in paracrine or autocrine fashion through specific cellular receptors.

More information

Title: NATURAL KILLER CELL FUNCTIONS AND SURFACE RECEPTORS

Title: NATURAL KILLER CELL FUNCTIONS AND SURFACE RECEPTORS LECTURE: 14 Title: NATURAL KILLER CELL FUNCTIONS AND SURFACE RECEPTORS LEARNING OBJECTIVES: The student should be able to: Describe the general morphology of the NK-cells. Enumerate the different functions

More information

Effector mechanisms of cell-mediated immunity: Properties of effector, memory and regulatory T cells

Effector mechanisms of cell-mediated immunity: Properties of effector, memory and regulatory T cells ICI Basic Immunology course Effector mechanisms of cell-mediated immunity: Properties of effector, memory and regulatory T cells Abul K. Abbas, MD UCSF Stages in the development of T cell responses: induction

More information

Tolerance 2. Regulatory T cells; why tolerance fails. Abul K. Abbas UCSF. FOCiS

Tolerance 2. Regulatory T cells; why tolerance fails. Abul K. Abbas UCSF. FOCiS 1 Tolerance 2. Regulatory T cells; why tolerance fails Abul K. Abbas UCSF FOCiS 2 Lecture outline Regulatory T cells: functions and clinical relevance Pathogenesis of autoimmunity: why selftolerance fails

More information

NKTR-214 plus NKTR-262, a Scientifically-Guided Rational Combination Approach for Immune Oncology

NKTR-214 plus NKTR-262, a Scientifically-Guided Rational Combination Approach for Immune Oncology plus NKTR-262, a Scientifically-Guided Rational Combination Approach for Immune Oncology Jonathan Zalevsky SVP, Biology and Preclinical Development Nektar Therapeutics World Preclinical Congress, 2017

More information

Immunology Lecture 4. Clinical Relevance of the Immune System

Immunology Lecture 4. Clinical Relevance of the Immune System Immunology Lecture 4 The Well Patient: How innate and adaptive immune responses maintain health - 13, pg 169-181, 191-195. Immune Deficiency - 15 Autoimmunity - 16 Transplantation - 17, pg 260-270 Tumor

More information

Immunology Basics Relevant to Cancer Immunotherapy: T Cell Activation, Costimulation, and Effector T Cells

Immunology Basics Relevant to Cancer Immunotherapy: T Cell Activation, Costimulation, and Effector T Cells Immunology Basics Relevant to Cancer Immunotherapy: T Cell Activation, Costimulation, and Effector T Cells Andrew H. Lichtman, M.D. Ph.D. Department of Pathology Brigham and Women s Hospital and Harvard

More information

The Adaptive Immune Responses

The Adaptive Immune Responses The Adaptive Immune Responses The two arms of the immune responses are; 1) the cell mediated, and 2) the humoral responses. In this chapter we will discuss the two responses in detail and we will start

More information

IMMUNOTHERAPY FOR CANCER A NEW HORIZON. Ekaterini Boleti MD, PhD, FRCP Consultant in Medical Oncology Royal Free London NHS Foundation Trust

IMMUNOTHERAPY FOR CANCER A NEW HORIZON. Ekaterini Boleti MD, PhD, FRCP Consultant in Medical Oncology Royal Free London NHS Foundation Trust IMMUNOTHERAPY FOR CANCER A NEW HORIZON Ekaterini Boleti MD, PhD, FRCP Consultant in Medical Oncology Royal Free London NHS Foundation Trust ASCO Names Advance of the Year: Cancer Immunotherapy No recent

More information

5/1/13. The proportion of thymus that produces T cells decreases with age. The cellular organization of the thymus

5/1/13. The proportion of thymus that produces T cells decreases with age. The cellular organization of the thymus T cell precursors migrate from the bone marrow via the blood to the thymus to mature 1 2 The cellular organization of the thymus The proportion of thymus that produces T cells decreases with age 3 4 1

More information

TCR, MHC and coreceptors

TCR, MHC and coreceptors Cooperation In Immune Responses Antigen processing how peptides get into MHC Antigen processing involves the intracellular proteolytic generation of MHC binding proteins Protein antigens may be processed

More information

The development of T cells in the thymus

The development of T cells in the thymus T cells rearrange their receptors in the thymus whereas B cells do so in the bone marrow. The development of T cells in the thymus The lobular/cellular organization of the thymus Immature cells are called

More information

C. Incorrect! MHC class I molecules are not involved in the process of bridging in ADCC.

C. Incorrect! MHC class I molecules are not involved in the process of bridging in ADCC. Immunology - Problem Drill 13: T- Cell Mediated Immunity Question No. 1 of 10 1. During Antibody-dependent cell mediated cytotoxicity (ADCC), the antibody acts like a bridge between the specific antigen

More information

Determinants of Immunogenicity and Tolerance. Abul K. Abbas, MD Department of Pathology University of California San Francisco

Determinants of Immunogenicity and Tolerance. Abul K. Abbas, MD Department of Pathology University of California San Francisco Determinants of Immunogenicity and Tolerance Abul K. Abbas, MD Department of Pathology University of California San Francisco EIP Symposium Feb 2016 Why do some people respond to therapeutic proteins?

More information

T Cell Activation. Patricia Fitzgerald-Bocarsly March 18, 2009

T Cell Activation. Patricia Fitzgerald-Bocarsly March 18, 2009 T Cell Activation Patricia Fitzgerald-Bocarsly March 18, 2009 Phases of Adaptive Immune Responses Phases of T cell responses IL-2 acts as an autocrine growth factor Fig. 11-11 Clonal Expansion of T cells

More information

T cell maturation. T-cell Maturation. What allows T cell maturation?

T cell maturation. T-cell Maturation. What allows T cell maturation? T-cell Maturation What allows T cell maturation? Direct contact with thymic epithelial cells Influence of thymic hormones Growth factors (cytokines, CSF) T cell maturation T cell progenitor DN DP SP 2ry

More information

Cell-mediated Immunity

Cell-mediated Immunity Cellular & Molecular Immunology Cell-mediated Immunity Nicholas M. Ponzio, Ph.D. Department of Pathology & Laboratory Medicine April 6, 2009 Today s Presentation: Overview Cellular Interactions In Humoral

More information

Tumors arise from accumulated genetic mutations. Tumor Immunology (Cancer)

Tumors arise from accumulated genetic mutations. Tumor Immunology (Cancer) Tumor Immunology (Cancer) Tumors arise from accumulated genetic mutations Robert Beatty MCB150 Mutations Usually have >6 mutations in both activation/growth factors and tumor suppressor genes. Types of

More information

Antigen Presentation and T Lymphocyte Activation. Abul K. Abbas UCSF. FOCiS

Antigen Presentation and T Lymphocyte Activation. Abul K. Abbas UCSF. FOCiS 1 Antigen Presentation and T Lymphocyte Activation Abul K. Abbas UCSF FOCiS 2 Lecture outline Dendritic cells and antigen presentation The role of the MHC T cell activation Costimulation, the B7:CD28 family

More information

08/02/59. Tumor Immunotherapy. Development of Tumor Vaccines. Types of Tumor Vaccines. Immunotherapy w/ Cytokine Gene-Transfected Tumor Cells

08/02/59. Tumor Immunotherapy. Development of Tumor Vaccines. Types of Tumor Vaccines. Immunotherapy w/ Cytokine Gene-Transfected Tumor Cells Tumor Immunotherapy Autologous virus Inactivation Inactivated virus Lymphopheresis Culture? Monocyte s Dendritic cells Immunization Autologous vaccine Development of Tumor Vaccines Types of Tumor Vaccines

More information

Medical Virology Immunology. Dr. Sameer Naji, MB, BCh, PhD (UK) Head of Basic Medical Sciences Dept. Faculty of Medicine The Hashemite University

Medical Virology Immunology. Dr. Sameer Naji, MB, BCh, PhD (UK) Head of Basic Medical Sciences Dept. Faculty of Medicine The Hashemite University Medical Virology Immunology Dr. Sameer Naji, MB, BCh, PhD (UK) Head of Basic Medical Sciences Dept. Faculty of Medicine The Hashemite University Human blood cells Phases of immune responses Microbe Naïve

More information

T Lymphocyte Activation and Costimulation. FOCiS. Lecture outline

T Lymphocyte Activation and Costimulation. FOCiS. Lecture outline 1 T Lymphocyte Activation and Costimulation Abul K. Abbas, MD UCSF FOCiS 2 Lecture outline T cell activation Costimulation, the B7:CD28 family Inhibitory receptors of T cells Targeting costimulators for

More information

Immunology for the Rheumatologist

Immunology for the Rheumatologist Immunology for the Rheumatologist Rheumatologists frequently deal with the immune system gone awry, rarely studying normal immunology. This program is an overview and discussion of the function of the

More information

Immunological Tolerance

Immunological Tolerance Immunological Tolerance Introduction Definition: Unresponsiveness to an antigen that is induced by exposure to that antigen Tolerogen = tolerogenic antigen = antigen that induces tolerance Important for

More information

Novel RCC Targets from Immuno-Oncology and Antibody-Drug Conjugates

Novel RCC Targets from Immuno-Oncology and Antibody-Drug Conjugates Novel RCC Targets from Immuno-Oncology and Antibody-Drug Conjugates Christopher Turner, MD Vice President, Clinical Science 04 November 2016 Uveal Melanoma Celldex Pipeline CANDIDATE INDICATION Preclinical

More information

Radiation Therapy as an Immunomodulator

Radiation Therapy as an Immunomodulator Radiation Therapy as an Immunomodulator Yvonne Mowery, MD, PhD February 20, 2017 Tumor/Immune System Balance Kalbasi, JCI 2013 UNC-Duke-NC State-Wake Forest Spring 2017 2 RT Can Shift Balance Toward Elimination

More information

The Adaptive Immune Response. B-cells

The Adaptive Immune Response. B-cells The Adaptive Immune Response B-cells The innate immune system provides immediate protection. The adaptive response takes time to develop and is antigen specific. Activation of B and T lymphocytes Naive

More information

Immunology - Lecture 2 Adaptive Immune System 1

Immunology - Lecture 2 Adaptive Immune System 1 Immunology - Lecture 2 Adaptive Immune System 1 Book chapters: Molecules of the Adaptive Immunity 6 Adaptive Cells and Organs 7 Generation of Immune Diversity Lymphocyte Antigen Receptors - 8 CD markers

More information

chapter 17: specific/adaptable defenses of the host: the immune response

chapter 17: specific/adaptable defenses of the host: the immune response chapter 17: specific/adaptable defenses of the host: the immune response defense against infection & illness body defenses innate/ non-specific adaptable/ specific epithelium, fever, inflammation, complement,

More information

Third line of Defense

Third line of Defense Chapter 15 Specific Immunity and Immunization Topics -3 rd of Defense - B cells - T cells - Specific Immunities Third line of Defense Specific immunity is a complex interaction of immune cells (leukocytes)

More information

Immune Regulation and Tolerance

Immune Regulation and Tolerance Immune Regulation and Tolerance Immunoregulation: A balance between activation and suppression of effector cells to achieve an efficient immune response without damaging the host. Activation (immunity)

More information

Innate immunity. Abul K. Abbas University of California San Francisco. FOCiS

Innate immunity. Abul K. Abbas University of California San Francisco. FOCiS 1 Innate immunity Abul K. Abbas University of California San Francisco FOCiS 2 Lecture outline Components of innate immunity Recognition of microbes and dead cells Toll Like Receptors NOD Like Receptors/Inflammasome

More information

Regulation of anti-tumor immunity through migration of immune cell subsets within the tumor microenvironment Thomas F. Gajewski, M.D., Ph.D.

Regulation of anti-tumor immunity through migration of immune cell subsets within the tumor microenvironment Thomas F. Gajewski, M.D., Ph.D. Regulation of anti-tumor immunity through migration of immune cell subsets within the tumor microenvironment Thomas F. Gajewski, M.D., Ph.D. Professor, Departments of Pathology and Medicine Program Leader,

More information

Basis of Immunology and

Basis of Immunology and Basis of Immunology and Immunophysiopathology of Infectious Diseases Jointly organized by Institut Pasteur in Ho Chi Minh City and Institut Pasteur with kind support from ANRS & Université Pierre et Marie

More information

Immune surveillance hypothesis (Macfarlane Burnet, 1950s)

Immune surveillance hypothesis (Macfarlane Burnet, 1950s) TUMOR-IMMUNITÄT A.K. Abbas, A.H. Lichtman, S. Pillai (6th edition, 2007) Cellular and Molecular Immunology Saunders Elsevier Chapter 17, immunity to tumors Immune surveillance hypothesis (Macfarlane Burnet,

More information

T Cell Receptor & T Cell Development

T Cell Receptor & T Cell Development T Cell Receptor & T Cell Development Questions for the next 2 lectures: How do you generate a diverse T cell population with functional TCR rearrangements? How do you generate a T cell population that

More information

T Cell Effector Mechanisms I: B cell Help & DTH

T Cell Effector Mechanisms I: B cell Help & DTH T Cell Effector Mechanisms I: B cell Help & DTH Ned Braunstein, MD The Major T Cell Subsets p56 lck + T cells γ δ ε ζ ζ p56 lck CD8+ T cells γ δ ε ζ ζ Cα Cβ Vα Vβ CD3 CD8 Cα Cβ Vα Vβ CD3 MHC II peptide

More information

Dr. Yi-chi M. Kong August 8, 2001 Benjamini. Ch. 19, Pgs Page 1 of 10 TRANSPLANTATION

Dr. Yi-chi M. Kong August 8, 2001 Benjamini. Ch. 19, Pgs Page 1 of 10 TRANSPLANTATION Benjamini. Ch. 19, Pgs 379-399 Page 1 of 10 TRANSPLANTATION I. KINDS OF GRAFTS II. RELATIONSHIPS BETWEEN DONOR AND RECIPIENT Benjamini. Ch. 19, Pgs 379-399 Page 2 of 10 II.GRAFT REJECTION IS IMMUNOLOGIC

More information

1. The scavenger receptor, CD36, functions as a coreceptor for which TLR? a. TLR ½ b. TLR 3 c. TLR 4 d. TLR 2/6

1. The scavenger receptor, CD36, functions as a coreceptor for which TLR? a. TLR ½ b. TLR 3 c. TLR 4 d. TLR 2/6 Allergy and Immunology Review Corner: Cellular and Molecular Immunology, 8th Edition By Abul K. Abbas, MBBS, Andrew H. H. Lichtman, MD, PhD and Shiv Pillai, MBBS, PhD. Chapter 4 (pages 62-74): Innate Immunity

More information

Immunity and Cancer. Doriana Fruci. Lab di Immuno-Oncologia

Immunity and Cancer. Doriana Fruci. Lab di Immuno-Oncologia Immunity and Cancer Doriana Fruci Lab di Immuno-Oncologia Immune System is a network of cells, tissues and organs that work together to defend the body against attacks of foreign invaders (pathogens, cancer

More information

New insights into CD8+ T cell function and regulation. Pam Ohashi Princess Margaret Cancer Centre

New insights into CD8+ T cell function and regulation. Pam Ohashi Princess Margaret Cancer Centre New insights into CD8+ T cell function and regulation Pam Ohashi Princess Margaret Cancer Centre New insights into CD8+ T cell function and regulation Pam Ohashi Princess Margaret Cancer Centre No Disclosures

More information

NATURAL KILLER T CELLS EBOOK

NATURAL KILLER T CELLS EBOOK 08 April, 2018 NATURAL KILLER T CELLS EBOOK Document Filetype: PDF 90.41 KB 0 NATURAL KILLER T CELLS EBOOK Natural killer T cells (NK T cells) are a type of lymphocyte, or white blood cell. Natural killer

More information

COURSE: Medical Microbiology, PAMB 650/720 - Fall 2008 Lecture 16

COURSE: Medical Microbiology, PAMB 650/720 - Fall 2008 Lecture 16 COURSE: Medical Microbiology, PAMB 650/720 - Fall 2008 Lecture 16 Tumor Immunology M. Nagarkatti Teaching Objectives: Introduction to Cancer Immunology Know the antigens expressed by cancer cells Understand

More information

University of Alberta

University of Alberta University of Alberta Regulation of the inhibitory receptor LIR-1 in human natural killer cells by Nicholas Lok-Tin Li A thesis submitted to the Faculty of Graduate Studies and Research in partial fulfillment

More information

Tolerance, autoimmunity and the pathogenesis of immunemediated inflammatory diseases. Abul K. Abbas UCSF

Tolerance, autoimmunity and the pathogenesis of immunemediated inflammatory diseases. Abul K. Abbas UCSF Tolerance, autoimmunity and the pathogenesis of immunemediated inflammatory diseases Abul K. Abbas UCSF Balancing lymphocyte activation and control Activation Effector T cells Tolerance Regulatory T cells

More information

The g c Family of Cytokines Prof. Warren J. Leonard M.D.

The g c Family of Cytokines Prof. Warren J. Leonard M.D. The Family of Cytokines Chief, Laboratory of Molecular Immunology Director, Immunology Center National Heart, Lung, and Blood Institute National Institutes of Health Department of Health and Human Services

More information

Chapter 13: Cytokines

Chapter 13: Cytokines Chapter 13: Cytokines Definition: secreted, low-molecular-weight proteins that regulate the nature, intensity and duration of the immune response by exerting a variety of effects on lymphocytes and/or

More information

CELL BIOLOGY - CLUTCH CH THE IMMUNE SYSTEM.

CELL BIOLOGY - CLUTCH CH THE IMMUNE SYSTEM. !! www.clutchprep.com CONCEPT: OVERVIEW OF HOST DEFENSES The human body contains three lines of against infectious agents (pathogens) 1. Mechanical and chemical boundaries (part of the innate immune system)

More information

Immunotherapy on the Horizon: Adoptive Cell Therapy

Immunotherapy on the Horizon: Adoptive Cell Therapy Immunotherapy on the Horizon: Adoptive Cell Therapy Joseph I. Clark, MD, FACP Professor of Medicine Loyola University Chicago Stritch School of Medicine Maywood, IL June 23, 2016 Conflicts of Interest

More information

Defensive mechanisms include :

Defensive mechanisms include : Acquired Immunity Defensive mechanisms include : 1) Innate immunity (Natural or Non specific) 2) Acquired immunity (Adaptive or Specific) Cell-mediated immunity Humoral immunity Two mechanisms 1) Humoral

More information

Tolerance 2. Regulatory T cells; why tolerance fails. FOCiS. Lecture outline. Regulatory T cells. Regulatory T cells: functions and clinical relevance

Tolerance 2. Regulatory T cells; why tolerance fails. FOCiS. Lecture outline. Regulatory T cells. Regulatory T cells: functions and clinical relevance 1 Tolerance 2. Regulatory T cells; why tolerance fails Abul K. Abbas UCSF FOCiS 2 Lecture outline Regulatory T cells: functions and clinical relevance Pathogenesis of autoimmunity: why selftolerance fails

More information

The Immune System. Innate. Adaptive. - skin, mucosal barriers - complement - neutrophils, NK cells, mast cells, basophils, eosinophils

The Immune System. Innate. Adaptive. - skin, mucosal barriers - complement - neutrophils, NK cells, mast cells, basophils, eosinophils Objectives - explain the rationale behind cellular adoptive immunotherapy - describe methods of improving cellular adoptive immunotherapy - identify mechanisms of tumor escape from cellular adoptive immunotherapy

More information

T Cell Development. Xuefang Cao, MD, PhD. November 3, 2015

T Cell Development. Xuefang Cao, MD, PhD. November 3, 2015 T Cell Development Xuefang Cao, MD, PhD November 3, 2015 Thymocytes in the cortex of the thymus Early thymocytes development Positive and negative selection Lineage commitment Exit from the thymus and

More information

Structure and Function of Antigen Recognition Molecules

Structure and Function of Antigen Recognition Molecules MICR2209 Structure and Function of Antigen Recognition Molecules Dr Allison Imrie allison.imrie@uwa.edu.au 1 Synopsis: In this lecture we will examine the major receptors used by cells of the innate and

More information

Dendritic Cell Based Immunotherapy for Cancer. Edgar G. Engleman, M.D.

Dendritic Cell Based Immunotherapy for Cancer. Edgar G. Engleman, M.D. Dendritic Cell Based Immunotherapy for Cancer Edgar G. Engleman, M.D. Two main DC subsets Myeloid (mydc) Derived from monocytes Capture/process/present Ag to T cells Activate NK cells and B cells Plasmacytoid

More information

NTD Vaccine Design Toolkit and Training Workshop Providence, RI January 05, 2011 Cytokines Leslie P. Cousens, PhD EpiVax, Inc.

NTD Vaccine Design Toolkit and Training Workshop Providence, RI January 05, 2011 Cytokines Leslie P. Cousens, PhD EpiVax, Inc. NTD Vaccine Design Toolkit and Training Workshop Providence, RI January 05, 2011 Cytokines Leslie P. Cousens, PhD EpiVax, Inc. Cytokines Properties of Cytokines Cytokines are proteins with specific roles

More information

Macrophage Activation & Cytokine Release. Dendritic Cells & Antigen Presentation. Neutrophils & Innate Defense

Macrophage Activation & Cytokine Release. Dendritic Cells & Antigen Presentation. Neutrophils & Innate Defense Macrophage Activation & Cytokine Release Dendritic Cells & Antigen Presentation Neutrophils & Innate Defense Neutrophils Polymorphonuclear cells (PMNs) are recruited to the site of infection where they

More information

Immunology. T-Lymphocytes. 16. Oktober 2014, Ruhr-Universität Bochum Karin Peters,

Immunology. T-Lymphocytes. 16. Oktober 2014, Ruhr-Universität Bochum Karin Peters, Immunology T-Lymphocytes 16. Oktober 2014, Ruhr-Universität Bochum Karin Peters, karin.peters@rub.de The role of T-effector cells in the immune response against microbes cellular immunity humoral immunity

More information

Immunobiology 7. The Humoral Immune Response

Immunobiology 7. The Humoral Immune Response Janeway Murphy Travers Walport Immunobiology 7 Chapter 9 The Humoral Immune Response Copyright Garland Science 2008 Tim Worbs Institute of Immunology Hannover Medical School 1 The course of a typical antibody

More information

1. Overview of Adaptive Immunity

1. Overview of Adaptive Immunity Chapter 17A: Adaptive Immunity Part I 1. Overview of Adaptive Immunity 2. T and B Cell Production 3. Antigens & Antigen Presentation 4. Helper T cells 1. Overview of Adaptive Immunity The Nature of Adaptive

More information

Enhancing the Clinical Activity of HER2/neu Specific T Cells. William Gwin, MD Internal Medicine, Resident University of Washington

Enhancing the Clinical Activity of HER2/neu Specific T Cells. William Gwin, MD Internal Medicine, Resident University of Washington Enhancing the Clinical Activity of HER2/neu Specific T Cells William Gwin, MD Internal Medicine, Resident University of Washington Immunotherapy and Cancer Cancer vaccines were originally used in melanoma

More information

Enhanced Cancer Vaccine Effectiveness with NKTR-214, a CD122-Biased Cytokine

Enhanced Cancer Vaccine Effectiveness with NKTR-214, a CD122-Biased Cytokine Enhanced Cancer Vaccine Effectiveness with NKTR-214, a CD122-Biased Cytokine Jonathan Zalevsky SVP, Biology and Preclinical Development Nektar Therapeutics SMI Cancer Vaccines, September 2017 Nektar Therapeutics

More information

CANCER IMMUNOPATHOLOGY. Eryati Darwin Faculty of Medicine Andalas University

CANCER IMMUNOPATHOLOGY. Eryati Darwin Faculty of Medicine Andalas University CANCER IMMUNOPATHOLOGY Eryati Darwin Faculty of Medicine Andalas University Padang 18 Mei 2013 INTRODUCTION Tumor: cells that continue to replicate, fail to differentiate into specialized cells, and become

More information

Objectives. Abbas Chapter 11: Immunological Tolerance. Question 1. Question 2. Question 3. Definitions

Objectives. Abbas Chapter 11: Immunological Tolerance. Question 1. Question 2. Question 3. Definitions Objectives Abbas Chapter 11: Immunological Tolerance Christina Ciaccio, MD Children s Mercy Hospitals and Clinics February 1, 2010 To introduce the concept of immunologic tolerance To understand what factors

More information

Effective activity of cytokine-induced killer cells against autologous metastatic melanoma including cells with stemness features

Effective activity of cytokine-induced killer cells against autologous metastatic melanoma including cells with stemness features Effective activity of cytokine-induced killer cells against autologous metastatic melanoma including cells with stemness features Loretta Gammaitoni, Lidia Giraudo, Valeria Leuci, et al. Clin Cancer Res

More information

T Cell Activation, Costimulation and Regulation

T Cell Activation, Costimulation and Regulation 1 T Cell Activation, Costimulation and Regulation Abul K. Abbas, MD University of California San Francisco 2 Lecture outline T cell antigen recognition and activation Costimulation, the B7:CD28 family

More information

Cellular Immune response. Jianzhong Chen, Ph.D Institute of immunology, ZJU

Cellular Immune response. Jianzhong Chen, Ph.D Institute of immunology, ZJU Cellular Immune response Jianzhong Chen, Ph.D Institute of immunology, ZJU Concept of adaptive immune response T cell-mediated adaptive immune response I. Concept of immune response A collective and coordinated

More information

7/6/2009. The study of the immune system and of diseases that occur as a result of inappropriate or inadequate actions of the immune system.

7/6/2009. The study of the immune system and of diseases that occur as a result of inappropriate or inadequate actions of the immune system. Diseases of Immunity 2009 CL Davis General Pathology Paul W. Snyder, DVM, PhD Purdue University Acknowledgements Pathologic Basis of Veterinary Disease, 4 th Ed Veterinary Immunology, An Introduction 8

More information

Mon, Wed, Fri 11:00 AM-12:00 PM. Owen, Judy, Jenni Punt, and Sharon Stranford Kuby-Immunology, 7th. Edition. W.H. Freeman and Co., New York.

Mon, Wed, Fri 11:00 AM-12:00 PM. Owen, Judy, Jenni Punt, and Sharon Stranford Kuby-Immunology, 7th. Edition. W.H. Freeman and Co., New York. Course Title: Course Number: Immunology Biol-341/541 Semester: Fall 2013 Location: HS 268 Time: Instructor: 8:00-9:30 AM Tue/Thur Dr. Colleen M. McDermott Office: Nursing Ed 101 (424-1217) E-mail*: mcdermot@uwosh.edu

More information

Cytokines (II) Dr. Aws Alshamsan Department of Pharmaceu5cs Office: AA87 Tel:

Cytokines (II) Dr. Aws Alshamsan Department of Pharmaceu5cs Office: AA87 Tel: Cytokines (II) Dr. Aws Alshamsan Department of Pharmaceu5cs Office: AA87 Tel: 4677363 aalshamsan@ksu.edu.sa Learning Objectives By the end of this lecture you will be able to: 1 Understand the physiological

More information

Third line of Defense. Topic 8 Specific Immunity (adaptive) (18) 3 rd Line = Prophylaxis via Immunization!

Third line of Defense. Topic 8 Specific Immunity (adaptive) (18) 3 rd Line = Prophylaxis via Immunization! Topic 8 Specific Immunity (adaptive) (18) Topics - 3 rd Line of Defense - B cells - T cells - Specific Immunities 1 3 rd Line = Prophylaxis via Immunization! (a) A painting of Edward Jenner depicts a cow

More information

Chapter 22: The Lymphatic System and Immunity

Chapter 22: The Lymphatic System and Immunity Bio40C schedule Lecture Immune system Lab Quiz 2 this week; bring a scantron! Study guide on my website (see lab assignments) Extra credit Critical thinking questions at end of chapters 5 pts/chapter Due

More information

Micr-6005, Current Concepts of Immunology (Rutgers course number: 16:681:543) Spring 2009 Semester

Micr-6005, Current Concepts of Immunology (Rutgers course number: 16:681:543) Spring 2009 Semester Micr-6005, Current Concepts of Immunology (Rutgers course number: 16:681:543) (3 Credits) Spring 2009 Semester Course Director: (732-235-4501, ) Please note that this course is offered once every 2 years.

More information

Immuno-Oncology Clinical Trials Update: Therapeutic Anti-Cancer Vaccines Issue 7 April 2017

Immuno-Oncology Clinical Trials Update: Therapeutic Anti-Cancer Vaccines Issue 7 April 2017 Delivering a Competitive Intelligence Advantage Immuno-Oncology Clinical Trials Update: Therapeutic Anti-Cancer Vaccines Issue 7 April 2017 Immuno-Oncology CLINICAL TRIALS UPDATE The goal of this MONTHLY

More information

TITLE: Development of Antigen Presenting Cells for adoptive immunotherapy in prostate cancer

TITLE: Development of Antigen Presenting Cells for adoptive immunotherapy in prostate cancer AD Award Number: W8-XWH-5-- TITLE: Development of Antigen Presenting Cells for adoptive immunotherapy in prostate cancer PRINCIPAL INVESTIGATOR: Mathias Oelke, Ph.D. CONTRACTING ORGANIZATION: Johns Hopkins

More information

Advances in Cancer Immunotherapy

Advances in Cancer Immunotherapy Advances in Cancer Immunotherapy Immunology 101 for the Non-Immunologist Arnold H. Zea, PhD azea@lsuhsc.edu Disclosures No relevant financial relationships to disclose This presentation does not contain

More information